Skip to main content
The Journal of Molecular Diagnostics : JMD logoLink to The Journal of Molecular Diagnostics : JMD
. 2024 Nov;26(11):952–961. doi: 10.1016/j.jmoldx.2024.07.001

The Correlation between Plasma Circulating Tumor DNA and Radiographic Tumor Burden

Evan M Alexander , Hunter A Miller , Michael E Egger †,, Melissa L Smith ‡,§,, Kavitha Yaddanapudi ‡,¶,, Mark W Linder ∗,‡,
PMCID: PMC11524323  PMID: 39181324

Abstract

Conventional blood-based biomarkers and radiographic imaging are excellent for use in monitoring different aspects of malignant disease, but given their specific shortcomings, their integration with other, complementary markers such as plasma circulating tumor DNA (ctDNA) will be beneficial toward a precision medicine–driven future. Plasma ctDNA analysis utilizes the measurement of cancer-specific molecular alterations in a variety of bodily fluids released by dying tumor cells to monitor and profile response to therapy, and is being employed in several clinical scenarios. Plasma concentrations of ctDNA have been reported to correlate with tumor burden. However, the strength of this association is generally poor and highly variable, confounding the interpretation of longitudinal plasma ctDNA measurements in conjunction with routine radiographic assessments. Herein is discussed what is currently understood with respect to the fundamental characteristics of tumor growth that dictate plasma ctDNA concentrations, with a perspective on its interpretation in conjunction with radiographically determined tumor burden assessments.


Key Points.

  • Cellular turnover rate likely plays a significant role in circulating tumor DNA (ctDNA) production through cellular death rate.

  • The value of circulating tumor DNA as a biomarker for disease surveillance relies upon its relationship to proliferation.

  • Longitudinal monitoring of ctDNA in cancer patients can provide valuable insight into tumor cell turnover and proliferation through direct measurement of changes in cell death rate.

Circulating tumor DNA (ctDNA) is a valuable clinical tool to support cancer care. Presently, plasma ctDNA analysis is being employed in several clinical scenarios, including monitoring for residual minimal disease in colorectal cancer,1 monitoring for the emergence of resistance mutations in patients with lung cancer,2 and serving as a surrogate DNA source for mutation profiling as an alternative to tumor tissue biopsy3 (ie, liquid biopsy). Kinetic changes in plasma ctDNA concentration ([ctDNA]) within the first weeks to months of therapeutic monitoring have demonstrated prognostic utility and are increasingly being applied to patient care.4, 5, 6, 7 In contrast, kinetic changes in plasma [ctDNA] in the later stages of therapeutic monitoring (once a patient is established on treatment) to detect progression is complicated by a lack of understanding about the relationship between plasma [ctDNA] and radiographically determined tumor burden measurements, which remain the standard of care for determining late-stage disease progression in solid tumors. Additionally, the use and interpretation of ctDNA measurements are further confounded given that a patient's radiologic assessments and blood draws for ctDNA detection, in practice and in the published literature, may not necessarily take place during the same visit. Herein is discussed what is currently understood with respect to the fundamental characteristics of tumor growth that dictate plasma [ctDNA], with a perspective on its interpretation in conjunction with radiographically determined tumor burden assessments. These reflections led to the conclusion that plasma [ctDNA] are primarily driven by a proliferating tumor burden. The implication of this conclusion supports the premise that sustained and increasing plasma [ctDNA] reflect clinical progression and suboptimal therapy prior to radiographically evident increases in tumor mass, and supports the application of longitudinal plasma [ctDNA] to aid in the early detection of disease progression and recurrence.8,9

More than 60% of patients diagnosed with nonmetastatic disease are at risk for eventual disease recurrence.10 The risk for recurrence depends on cancer type and can range from approximately 17% in colorectal cancer, to >75% in glioblastoma.11, 12, 13 Furthermore, in >30% of patients with advanced melanoma, the disease ultimately progresses despite first-line therapy, and 52% of patients with stage III melanoma may experience recurrence despite undergoing adjuvant therapy.14, 15, 16, 17 Although certain risk factors can be used to identify patients whose disease is more likely to recur or progress, many patients who present with recurrence are asymptomatic.18,19 Currently, radiographic imaging is the clinical tool most universally used to assess solid tumor progression.20 Response Evaluation Criteria in Solid Tumors (RECIST; version 1.1) guidelines have been established to standardize the criteria for determining whether a given tumor is responding, stable, or progressing, depending on a change in size as determined by cross-sectional imaging. RECIST criteria are applied to radiographic measurements by computed tomography or positron emission tomography to determine whether the patient shows complete response (<10 mm all lesions), partial response (30% decrease in sum diameter), progressive disease (20% increase in sum diameter), or stable disease (insignificant size change).20 Positron emission tomography–based radiography increases sensitivity by using an 18F-fluorodeoxyglucose substrate to evaluate metabolically active tumor cells.21,22 However, radiographic surveillance has limitations, including low frequency of monitoring, inability to monitor longitudinal molecular heterogeneity, patient exposure to ionizing radiation, and a low sensitivity in recurrence detection.23, 24, 25

Cell-free DNA (cfDNA), and its tumor constituent, ctDNA, has gained traction in the literature on clinical diagnostics and as a tool in clinical trials over the past decade.26, 27, 28, 29, 30, 31, 32 This liquid-biopsy approach utilizes the measurement of cancer-specific molecular alterations in a variety of bodily fluids released by dying tumor cells to monitor and profile the response to therapy. Research has shown that ctDNA quantities, expressed either as a percentage of variant-allele frequency or as the concentration of copies per milliliter of body fluid, are associated with prognosis and overall survival.33, 34, 35, 36, 37 However, ctDNA quantities have also shown inconsistencies in relation to other blood-based biomarkers and radiographic imaging.38, 39, 40, 41 Currently, a lack of understanding about the basis of any correlation between ctDNA and other well-established diagnostic techniques may be stifling its incorporation into widespread clinical use. It is argued that the underlying factor dictating the relationship between plasma ctDNA and tumor burden is tumor proliferation.

ctDNA Origins and Kinetics

cfDNA is a collective term used to describe small DNA fragments that circulate freely in bodily fluids, and may be of nuclear or mitochondrial origin.31 These fragments originate largely from necrosis, apoptosis, and cellular turnover of hematopoietic cells.42, 43, 44, 45, 46 As cancer cells die, cfDNA containing tumor-specific genetic alterations can enter the systemic circulation and is detected as ctDNA.31,32,47 ctDNA is observed either in its free form, associated with single histones, within larger nucleosome complexes, or within microparticles such as exosomes and apoptotic bodies.31,48 ctDNA is rapidly eliminated in vivo, with a median half-life (t1/2) of 35 minutes.49

The fact that apoptosis and or necrosis of tumor tissue is the source of plasma ctDNA, and that plasma ctDNA is rapidly eliminated, would lead to the expectation that patients with elevated concentrations of plasma ctDNA would demonstrate reductions in tumor mass. However, in practice, increasing tumor burden is typically associated with increasing plasma [ctDNA],50 but the correlation between tumor burden and ctDNA is poor to moderate, with correlation coefficients on the order of (r2 ≈ 0.5). This poor degree of correlation limits the utility of plasma ctDNA measurements used for predicting tumor burden and confounds the interpretation of plasma ctDNA measurements. In recent work it was observed that the ratio of plasma [ctDNA] to tumor burden was approximately 5-fold greater, with a significant correlation with tumor burden (r2 = 0.91) under conditions of worsening disease.51 These data led to the exploration of the underlying factors that influence the relationship between plasma ctDNA and tumor burden.

Unless otherwise specified by the study design, most plasma ctDNA measurements published in the literature, and in particular those measurements obtained during maintenance systemic therapy, represent what can be considered a random steady-state measurement. Pharmacokinetic principles dictate that in order to maintain steady-state plasma concentrations of a substance with rapid elimination, a corresponding rate of infusion is required.

The following example is provided to illustrate the conditions necessary for sustaining elevated plasma concentrations of ctDNA. Assuming a volume of distribution (Vd) of 5 L (blood compartment) and a median t1/2 of ctDNA (0.58 hours) from lung cancer patients after tumor resection observed by Chen,49 the clearance (CL) of ctDNA can be approximated, using Equation 1, as 5.96 L/hour.

CL=0.692Vd/t1/2 (1)

For ctDNA to maintain steady-state concentration (Css), a compensatory infusion rate from the tumor must also be present (Equation 2).

Css=Infusionrate/Eliminationrate (2)

Because tumor cell death is likely the primary origin of ctDNA,28,30,52 the infusion rate must be proportional to the tumor cell death rate (Equation 3). (Note: Here, it is assumed that the amount of ctDNA released by living cells and circulating tumor cells is negligible). Thus, if ctDNA is measurable, then there must be a proportional amount of continual cell death occurring in the tumor:

Css=xDeathrate/Eliminationrate (3)

where x represents the combined effects of other factors, such as ctDNA released by living tumor cells and circulating tumor cells, DNA shedding probability as described by Avanzini et al,53 and ctDNA that does not escape the tumor microenvironment due to the phagocytosis of apoptotic bodies by neighboring cells and macrophages.54 These factors are still not fully understood; therefore, x = 1 is assumed for simplicity.

The range of plasma ctDNA (copies/mL) observed by Bettegowda et al55 among various cancer types was 1 to 200,000 copies/mL. Using Equation 3, sustaining a steady-state concentration of 1 copy/mL of ctDNA would necessitate a cell death rate of 24,000 cells/day. Late-stage colorectal cancer in which plasma [ctDNA] may exceed 20,000 copies/mL would suggest a cell death rate of 4.8 × 108 cells/day. A tissue volume of 1 cm3 is approximately 1 × 108 to 1 × 109 cells.56 This indicates that the cell death rate would result in a decrease in tumor size of 0.48 to 4.8 cm3/day. Paradoxically, patients with late-stage cancer, metastasis, and elevated plasma [ctDNA] frequently exhibit increasing tumor burden on radiography.

Life and Death

Cellular turnover is a crucial component of homeostasis in healthy tissue. The lifespan of human eukaryotic cells can vary from 5 days in the case of colon epithelial cells,57 to 100 days in erythrocytes,58 to 200 to 300 days in liver cells,59 and persisting indefinitely in the case of neuronal cells.60 Hematopoietic cells represent 85% of total cellular turnover per day57 while contributing nearly 85% of total cfDNA in healthy individuals.46 Furthermore, the cell types associated with cancers demonstrating the highest concentrations of ctDNA (gastrointestinal, epithelial) possess some of the highest proliferation rates in the body outside of hematopoietic cells.55,57

Considering this observation, the cellular turnover rate likely plays a significant role in cfDNA/ctDNA production through cell death rate, as described in Equation 4:

CellularturnoverrateProliferationrateDeathrate (4)

Although ctDNA may reflect cell death and, indirectly, cellular turnover, its value as a biomarker in disease surveillance relies upon its relationship to proliferation. Dysregulation between proliferation and apoptosis is the driving force of tumorigenesis.61 However, complete ablation of apoptosis with uncontrolled proliferation would lead to a tumor volume of 1 cm3 (approximately 1 × 109 cells), increasing to 1000 cm3 (a tumor incompatible with life) within 10 cell-doubling events.62 Thus, continual cell death must play a role in an actively proliferating tumor in order to maintain sustainable growth.

Tumor cell death via apoptosis and necrosis have been identified as the most prominent sources of ctDNA.28,30,52 In 2001, Liu et al63 quantified proliferation and apoptosis in breast cancer biopsy samples and found that increased apoptotic cell counts were strongly associated with tumor grade, size, metastasis, and shortened disease-specific survival. Similar observations have been reported in colorectal cancer,64 endometrial adenocarcinoma,65 and prostate cancer.66 Apoptosis-induced proliferation is an established link wherein apoptotic cells produce and secrete a burst of mitogens, signaling proliferation in neighboring cells.67, 68, 69 Passive cell death through necrosis may play a smaller, but important, role in proliferation signaling via the induction and enhancement of apoptosis.70, 71, 72 The understanding of the mechanisms and pathways of cell death has increased greatly in the past two decades.73 An intermediary between apoptosis and necrosis—necroptosis, wherein cells undergoing apoptosis, remain undead, and continually secrete mitogens—may play a significant role in tumor proliferation and metastasis.68,74,75 The significance and influence that lesser established death mechanisms (pyroptosis, ferroptosis, autosis, parthanatos) have on tumorigenesis and ctDNA release kinetics are currently unknown.

ctDNA, Tumor Burden, and Disease Progression

Using a simple generalization, an increase in the proliferation rate of malignant cells enables disease progression. To facilitate the earliest possible detection of progression or recurrence, then, requires the earliest possible detection of a change in proliferation rate. Highly sensitive, direct measurement of in vivo tumor proliferation rate is currently not achievable. Additionally, radiographic tumor burden size is not a direct measure of proliferation rate.

Figure 1 is a schematic representation of how plasma concentrations of ctDNA are influenced by the various stages of tumor development, therapeutic response, and recurrence.

Figure 1.

Figure 1

A model of tumor progression and ctDNA release. Panel 1: At the initiation stage of tumor proliferation, growth rate predominates, with minimal cell death, resulting in ctDNA concentrations ([ctDNA]) that are less than the lower limit of detection (LOD). Tumor proliferation leads to the formation of a nutrient-deprived necrotic core (gray cells). Panel 2: Cancer cells proliferate and increase in abundance. Cell death rate increases and low [ctDNA] may be detectable at this stage (brown region). Panel 3: At later stages of disease, the tumor microenvironment is fully established, and cell death rate increases dramatically. Increased cellular turnover allows the opportunity for the malignancy to differentiate and become heterogenous as invasive cancer cells appear (darkred regions). Panel 4: The initiation of therapy promotes tumor cell death and diminishes cellular proliferation, leading to rapid clearance of ctDNA due to the absence of proliferation. Panel 5: Residual proliferation after therapy allows for tumor subclones to reestablish themselves, including new therapy-resistant cells. Panel 6: New therapy-resistant subclones begin yielding ctDNA as proliferation increases (green region).

Tumor burden is maintained by the balance between proliferation and cell death (Equation 5).

ΔTBΔrgΔrd (5)

where TB is the tumor burden, rg is the cell proliferation rate, and rd is the cell death rate.

This relationship is illustrated in Figure 2, where relative tumor proliferation rates can be inferred at various stages after treatment, and the cell death rate is represented by [ctDNA]. In a scenario of increasing tumor burden, the proliferation rate exceeds the cell death rate. A clinical scenario in which the proliferation rate is increasing with a proportionate increase in cell death rate would result in no change in tumor mass, and radiographic tumor burden would fail to detect progression. This scenario would appear as stable disease in RECIST, despite increasing proliferation.

Figure 2.

Figure 2

Two theoretical scenarios—disease recurrence and complete therapeutic response—comparing the changes in the rates of tumor proliferation and cell death (represented by ctDNA concentration [ctDNA]), both in arbitrary units. Given the relationship ΔTB ∝ Δrg – Δrd ([ctDNA]), where TB is the tumor burden, rg is the proliferation rate, and rd is the cell death rate, the relative tumor burden can be inferred at various stages after treatment and during disease surveillance, based on the relative rates of tumor proliferation and [ctDNA] release. Arrows indicate the time points relevant in the calculation of Δ. The theoretical lower limit of detection (LOD) of ctDNA is represented by the red line; theoretical radiographic LOD is represented by the blue line. A: Partial response with recurrence. Prior to treatment, proliferation is increased (Δrg = 20), resulting in an increased rate of cell death (Δrd = 5). The net effect is an increase in ctDNA, with a net increase in TB of 15 after 1 month. (A1): At 1 month after treatment, proliferation is partially reduced (Δrg = −40), resulting in a reduction in cell death rate (Δrd = −30), driving decreases in both TB (ΔTB = −10) and plasma [ctDNA]. (A2): Several months after treatment, proliferation remains reduced (Δrg = 0) and the cell death rate normalizes to the proliferation rate (Δrd = 0), resulting in persistently low but detectable quantities of plasma ctDNA without any change in TB (ΔTB = 0) relative to the previous month. (A3): Developing resistance to therapy allows for an increased rate of proliferation (Δrg = 20), which drives an increase in cell death rate (Δrd = 15), resulting in an acute increase in plasma ctDNA and a gradual increase in TB (ΔTB = 5) relative to the previous month. B: Complete response. As in A, prior to treatment, proliferation increases (Δrg = 20), resulting in an increased rate of cell death (Δrd = 5). The net effect is an increase in ctDNA, with an increase in TB of 15 after 1 month. (B1): At 1 month after treatment, proliferation is dramatically reduced (Δrg = −110), resulting in a reduction in cell death rate (Δrd = −70), such that plasma [ctDNA] are below the LOD, and TB is also reduced (ΔTB = −40). (B2): Several months after treatment, the rates of proliferation and cell death remain unchanged, resulting in persistently undetectable plasma [ctDNA] and no change in TB relative to the previous month. (B3): Complete response ablates proliferation (Δrg = −20), and death rate persists (Δrd = 0), resulting in continued concentrations of plasma ctDNA below the LOD and a reduction in residual TB (ΔTB = −20) relative to the previous month.

In the scenario of the misidentification of an increased proliferation rate as stable disease, ctDNA would help to elucidate the previously undetectable change in cellular turnover and proliferation due to increasing [ctDNA] caused by an increasing cell death rate. This was observed clinically in patients with colorectal cancer, in whom increases in plasma [ctDNA] preceded radiographically identified progressive disease by up to 11.5 months.76, 77, 78 In patients with esophageal adenocarcinoma and non–small-cell lung cancer, increases in plasma ctDNA preceded radiographically identified progressive disease by 100 days and a median of 88 days, respectively.79,80 An additional beneficial aspect of ctDNA is in predicting overall disease-free survival after treatment based on plasma ctDNA clearance rate.81 ctDNA clearance is the net measure of infusion and elimination. Immediately after the initiation of therapy, [ctDNA] can show varied clearance kinetics based on therapy.81 For instance, after surgical intervention, the removal of bulk cells can lead to deceases in [ctDNA] of up to 99%.82,83 This decrease most likely reflects rapid elimination in the absence of a tumor tissue source. The initiation of radiation therapy or chemotherapy may lead to transient peaks in [ctDNA] in some patients within the first 72 hours, followed by a sharp decrease.84, 85, 86, 87 This scenario most likely reflects a rapid and transient infusion from dying cells, followed by the absence of continued infusion from the responding tumor. Patients can have varied responses to immunotherapy, with rapid clearance, fluctuations/rises within 3 to 16 weeks, or initial rapid clearance followed by a steady increase in concentration. Each of these scenarios would be representative of the net effect of therapy on the proliferation state of the tumor. A reduced plasma ctDNA clearance rate after treatment, and failure to completely ablate measurable ctDNA, are also indicative of poor progression-free survival.81 This is consistent with a scenario in which tumor proliferation capacity has not been fully eliminated. A complete response to therapy leads to sharp and complete decreases in ctDNA,49,83 which is consistent with the absence of tumor proliferation and rapid elimination of the released ctDNA. A partial response to therapy leads to incomplete or slow clearance of ctDNA, most likely the result of a partial reduction in tumor-proliferation rate, with no change in elimination. As established earlier, ctDNA is a component of cellular turnover and related to proliferation. Thus, residual ctDNA indicates residual tumor cellular turnover and proliferation after treatment, while the complete ablation of ctDNA indicates ablation of cellular turnover and proliferation.

Initially, one may associate decreasing tumor burden with increasing cell death rate and expect increased [ctDNA]; however, decreasing tumor burden indicates only that the cell death rate exceeds the proliferation rate; it cannot be used as a surrogate for an absolute value of either rate. Due to the rapid elimination (short t1/2) of ctDNA, complete response and decreasing tumor burden quickly deplete stores of DNA available for shedding into the circulation, the result being great reductions in both rates, with the cell death rate exceeding the proliferation rate, allowing for a decrease in tumor burden. If the ctDNA stores are not depleted after therapy, there must be a replenishing mechanism via residual cellular turnover and proliferation. This residual tumor activity lays the foundation for disease recurrence. Cell death and proliferation rates exist in an equilibrium that drives tumor dynamics and cellular turnover. Two theoretical patient scenarios, complete response and partial response (Figure 2), are further elaborated in the context of cellular turnover (Figure 3). A complete response to therapy yields a large reduction in tumor cellular turnover and longer disease-free survival (Figure 3), while a partial response to therapy maintains residual proliferation activity despite an initial reduction in tumor burden (Figure 3).

Figure 3.

Figure 3

Graphical representation of the relationship between the rates of cell proliferation and death in the context of changes in tumor burden and tumor cell turnover. A: The diagonal line represents stable tumor burden as the rates of cell proliferation and death increase proportionately with each other. In the blue region, the cell death rate is high, while the proliferation rate is low, which reduces tumor burden. In the red region, the proliferation rate is high, while the cell death rate is low, which increases tumor burden. The shaded arcs represent zones of cellular turnover that are low (bottom left) and high (top right). The numbered points represent longitudinal radiologic measurements of disease status. B: 1 → 2, Disease burden remains stable despite an increased proliferation rate (Δrg = Δrd [ctDNA]; increasing in parallel); 2 → 3, the proliferation rate begins to surpass the cell death rate, resulting in increased radiographic tumor burden (Δrg > Δrd); 3 → 4, a complete response to therapy greatly reduces the rate of tumor cell turnover, resulting in a steep decrease in the rates of cell proliferation and death [Δrg < Δrd; Δrd < lower limit of detection (LOD)]. C: 1 → 3, Clinical progression identical to that in the previous case; 3 → 4, a partial response to therapy leads to residual proliferation and continued cellular turnover (Δrg < Δrd; Δrd > LOD); 4 → 5, continued cellular turnover generates subpopulations of highly proliferative cells, leading to tumor recurrence (Δrg > Δrd). Theoretical ctDNA LOD is represented by the red line; theoretical radiographic LOD is represented by the blue line.

Conclusion

Due to its rapid clearance, ctDNA represents a continuously updating snapshot of the rate of tumor cell death. Longitudinal monitoring of ctDNA in cancer patients can provide valuable insight into tumor cell turnover (activity) and proliferation through direct measurement of changes in the cell death rate. Theoretically, stable tumor burden with detectable ctDNA suggests proliferation in excess of death and an increased risk for recurrence; increasing ctDNA with stable tumor burden indicates a parallel increase in death rate and proliferation rate, signifying imminent recurrence; and stable tumor burden with no detectable ctDNA indicates minimal cellular turnover and a low risk for recurrence. Conventional blood-based biomarkers and radiographic imaging are excellent for monitoring the aspects of disease, but each has specific shortcomings that prevent full integration into a precision medicine–driven future. Despite inconsistent correlation with various cancer markers and variations in levels detected among different tumor types, changes in ctDNA quantities frequently precede changes in radiographically determined tumor burden and methods of cancer biomarker surveillance when used to predict or detect disease recurrence. This facet may be attributed to ctDNA's unique representation of the balance between proliferation and cell death, which can result in increased sensitivity in detection compared to radiographic imaging. However, as with other blood-based biomarkers, ctDNA presents challenges regarding reproducibility and harmonization, which may limit widespread adoption by clinics.88, 89, 90, 91 When tumors are small, the signal-to-noise ratio is low due to the presence of a high background made of wild-type circulating DNA, which can limit the analytical sensitivity of the assay.89 Biological factors, such as differences in the degree and rate of association or disassociation with various intracellular and extracellular components, may cause ctDNA to be cleared from the circulation at rates that differ between patients.91 Additionally, pre-analytical discrepancies in biospecimen collection, processing, and storage practices are an ongoing challenge that prohibits widespread adoption by clinics.90 Even so, ctDNA provides substantial technical value to augment radiographic surveillance techniques. The theoretical lower limit of detection of ctDNA, which is limited by cell death events and pharmacokinetic parameters, is multiple orders of magnitude lower than tumor burden, which is currently confined to millimeter-based measurements. Temporal resolution of surveillance is also improved with the use of a blood-based biomarker, which may lessen the time required for detecting recurrence. In the coming decade, ctDNA is poised to become a widely used, powerful surveillance tool that aides in the understanding and early detection of tumor recurrence and therapeutic resistance.

Disclosure Statement

None declared.

Footnotes

Part of this work was performed with the assistance of the Center for Cancer Immunology and Immunotherapy (CCII), which is supported by NIH grant P20GM135004 (KY IDeA Networks of Biomedical Research Excellence) and the University of Louisville Health–Brown Cancer Center (M.W.L., M.E.E., K.Y., and M.L.S.).

References

  • 1.Tie J., Wang Y., Tomasetti C., Li L., Springer S., Kinde I., Silliman N., Tacey M., Wong H.L., Christie M., Kosmider S., Skinner I., Wong R., Steel M., Tran B., Desai J., Jones I., Haydon A., Hayes T., Price T.J., Strausberg R.L., Diaz L.A., Jr., Papadopoulos N., Kinzler K.W., Vogelstein B., Gibbs P. Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer. Sci Transl Med. 2016;8 doi: 10.1126/scitranslmed.aaf6219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Horn L., Whisenant J.G., Wakelee H., Reckamp K.L., Qiao H., Leal T.A., Du L., Hernandez J., Huang V., Blumenschein G.R., Waqar S.N., Patel S.P., Nieva J., Oxnard G.R., Sanborn R.E., Shaffer T., Garg K., Holzhausen A., Harrow K., Liang C., Lim L.P., Li M., Lovly C.M. Monitoring therapeutic response and resistance: analysis of circulating tumor DNA in patients with ALK+ lung cancer. J Thorac Oncol. 2019;14:1901–1911. doi: 10.1016/j.jtho.2019.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Diaz L.A., Jr., Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol. 2014;32:579–586. doi: 10.1200/JCO.2012.45.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Pazdirek F., Minarik M., Benesova L., Halkova T., Belsanova B., Macek M., Stepanek L., Hoch J. Monitoring of early changes of circulating tumor DNA in the plasma of rectal cancer patients receiving neoadjuvant concomitant chemoradiotherapy: evaluation for prognosis and prediction of therapeutic response. Front Oncol. 2020;10:1028. doi: 10.3389/fonc.2020.01028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Cheng M.L., Lau C.J., Milan M.S.D., Supplee J.G., Riess J.W., Bradbury P.A., Janne P.A., Oxnard G.R., Paweletz C.P. Plasma ctDNA response is an early marker of treatment effect in advanced NSCLC. JCO Precis Oncol. 2021;5 doi: 10.1200/PO.20.00419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Gray E.S., Rizos H., Reid A.L., Boyd S.C., Pereira M.R., Lo J., Tembe V., Freeman J., Lee J.H., Scolyer R.A., Siew K., Lomma C., Cooper A., Khattak M.A., Meniawy T.M., Long G.V., Carlino M.S., Millward M., Ziman M. Circulating tumor DNA to monitor treatment response and detect acquired resistance in patients with metastatic melanoma. Oncotarget. 2015;6:42008–42018. doi: 10.18632/oncotarget.5788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Braune J., Keller L., Schiller F., Graf E., Rafei-Shamsabadi D., Wehrle J., Follo M., Philipp U., Hussung S., Pfeifer D., Mix M., Duyster J., Fritsch R., von Bubnoff D., Meiss F., von Bubnoff N. Circulating tumor DNA allows early treatment monitoring in BRAF- and NRAS-mutant malignant melanoma. JCO Precis Oncol. 2020;4:20–31. doi: 10.1200/PO.19.00174. [DOI] [PubMed] [Google Scholar]
  • 8.Chang G.A., Tadepalli J.S., Shao Y., Zhang Y., Weiss S., Robinson E., Spittle C., Furtado M., Shelton D.N., Karlin-Neumann G., Pavlick A., Osman I., Polsky D. Sensitivity of plasma BRAFmutant and NRASmutant cell-free DNA assays to detect metastatic melanoma in patients with low RECIST scores and non-RECIST disease progression. Mol Oncol. 2016;10:157–165. doi: 10.1016/j.molonc.2015.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Gouda M.A., Huang H.J., Piha-Paul S.A., Call S.G., Karp D.D., Fu S., Naing A., Subbiah V., Pant S., Dustin D.J., Tsimberidou A.M., Hong D.S., Rodon J., Meric-Bernstam F., Janku F. Longitudinal monitoring of circulating tumor DNA to predict treatment outcomes in advanced cancers. JCO Precis Oncol. 2022;6 doi: 10.1200/PO.21.00512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Luigjes-Huizer Y.L., Tauber N.M., Humphris G., Kasparian N.A., Lam W.W.T., Lebel S., Simard S., Smith A.B., Zachariae R., Afiyanti Y., Bell K.J.L., Custers J.A.E., de Wit N.J., Fisher P.L., Galica J., Garland S.N., Helsper C.W., Jeppesen M.M., Liu J., Mititelu R., Monninkhof E.M., Russell L., Savard J., Speckens A.E.M., van Helmondt S.J., Vatandoust S., Zdenkowski N., van der Lee M.L. What is the prevalence of fear of cancer recurrence in cancer survivors and patients? A systematic review and individual participant data meta-analysis. Psycho Oncol. 2022;31:879–892. doi: 10.1002/pon.5921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Mahvi D.A., Liu R., Grinstaff M.W., Colson Y.L., Raut C.P. Local cancer recurrence: the realities, challenges, and opportunities for new therapies. CA A Cancer J Clin. 2018;68:488–505. doi: 10.3322/caac.21498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Nabors L.B., Portnow J., Ahluwalia M., Baehring J., Brem H., Brem S., Butowski N., Campian J.L., Clark S.W., Fabiano A.J., Forsyth P., Hattangadi-Guth J., Holdhoff M., Horbinski C., Junck L., Kaley T., Kumthekar P., Loeffler J.S., Mrugala M.M., Nagpal S., Pandey M., Parney I., Peters K., Puduvalli V.K., Robins I., Rockhill J., Rusthoven C., Shonka N., Shrieve D.C., Swinnen L.J., Weiss S., Wen P.Y., Willmarth N.E., Bergman M.A., Darlow S.D. Central nervous system cancers, version 3.2020, NCCN clinical practice guidelines in oncology. J Natl Compr Cancer Netw. 2020;18:1537–1570. doi: 10.6004/jnccn.2020.0052. [DOI] [PubMed] [Google Scholar]
  • 13.Pugh S.A., Shinkins B., Fuller A., Mellor J., Mant D., Primrose J.N. Site and stage of colorectal cancer influence the likelihood and distribution of disease recurrence and postrecurrence survival: data from the FACS randomized controlled trial. Ann Surg. 2016;263:1143–1147. doi: 10.1097/SLA.0000000000001351. [DOI] [PubMed] [Google Scholar]
  • 14.Cowey C.L., Liu F.X., Boyd M., Aguilar K.M., Krepler C. Real-world treatment patterns and clinical outcomes among patients with advanced melanoma: a retrospective, community oncology-based cohort study (a STROBE-compliant article) Medicine. 2019;98 doi: 10.1097/MD.0000000000016328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Lee A.Y., Droppelmann N., Panageas K.S., Zhou Q., Ariyan C.E., Brady M.S., Chapman P.B., Coit D.G. Patterns and timing of initial relapse in pathologic stage II melanoma patients. Ann Surg Oncol. 2017;24:939–946. doi: 10.1245/s10434-016-5642-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Romano E., Scordo M., Dusza S.W., Coit D.G., Chapman P.B. Site and timing of first relapse in stage III melanoma patients: implications for follow-up guidelines. J Clin Oncol. 2010;28:3042–3047. doi: 10.1200/JCO.2009.26.2063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kluger H.M., Tawbi H.A., Ascierto M.L., Bowden M., Callahan M.K., Cha E., Chen H.X., Drake C.G., Feltquate D.M., Ferris R.L., Gulley J.L., Gupta S., Humphrey R.W., LaVallee T.M., Le D.T., Hubbard-Lucey V.M., Papadimitrakopoulou V.A., Postow M.A., Rubin E.H., Sharon E., Taube J.M., Topalian S.L., Zappasodi R., Sznol M., Sullivan R.J. Defining tumor resistance to PD-1 pathway blockade: recommendations from the first meeting of the SITC immunotherapy resistance taskforce. J Immunother Cancer. 2020;8 doi: 10.1136/jitc-2019-000398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Park T.S., Phan G.Q., Yang J.C., Kammula U., Hughes M.S., Trebska-McGowan K., Morton K.E., White D.E., Rosenberg S.A., Sherry R.M. Routine computer tomography imaging for the detection of recurrences in high-risk melanoma patients. Ann Surg Oncol. 2017;24:947–951. doi: 10.1245/s10434-017-5768-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Schadendorf D., Long G.V., Stroiakovski D., Karaszewska B., Hauschild A., Levchenko E., Chiarion-Sileni V., Schachter J., Garbe C., Dutriaux C., Gogas H., Mandala M., Haanen J.B.A.G., Lebbe C., Mackiewicz A., Rutkowski P., Grob J.-J., Nathan P., Ribas A., Davies M.A., Zhang Y., Kaper M., Mookerjee B., Legos J.J., Flaherty K.T., Robert C. Three-year pooled analysis of factors associated with clinical outcomes across dabrafenib and trametinib combination therapy phase 3 randomised trials. Eur J Cancer. 2017;82:45–55. doi: 10.1016/j.ejca.2017.05.033. [DOI] [PubMed] [Google Scholar]
  • 20.Eisenhauer E.A., Therasse P., Bogaerts J., Schwartz L.H., Sargent D., Ford R., Dancey J., Arbuck S., Gwyther S., Mooney M., Rubinstein L., Shankar L., Dodd L., Kaplan R., Lacombe D., Verweij J. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45:228–247. doi: 10.1016/j.ejca.2008.10.026. [DOI] [PubMed] [Google Scholar]
  • 21.Min S.J., Jang H.J., Kim J.H. Comparison of the RECIST and PERCIST criteria in solid tumors: a pooled analysis and review. Oncotarget. 2016;7:27848–27854. doi: 10.18632/oncotarget.8425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Wahl R.L., Jacene H., Kasamon Y., Lodge M.A. From RECIST to PERCIST: evolving considerations for PET response criteria in solid tumors. J Nucl Med. 2009;50(Suppl 1):122S–150S. doi: 10.2967/jnumed.108.057307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Jeh S.K., Kim S.H., Kang B.J. Comparison of the diagnostic performance of response evaluation criteria in solid tumor 1.0 with response evaluation criteria in solid tumor 1.1 on MRI in advanced breast cancer response evaluation to neoadjuvant chemotherapy. Korean J Radiol. 2013;14:13–20. doi: 10.3348/kjr.2013.14.1.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Stewart C., Smith-Bindman R. It is time to inform patients of medical imaging risks. JAMA Netw Open. 2021;4 doi: 10.1001/jamanetworkopen.2021.29681. [DOI] [PubMed] [Google Scholar]
  • 25.Villaruz L.C., Socinski M.A. The clinical viewpoint: definitions, limitations of RECIST, practical considerations of measurement. Clin Cancer Res. 2013;19:2629–2636. doi: 10.1158/1078-0432.CCR-12-2935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Crowley E., Di Nicolantonio F., Loupakis F., Bardelli A. Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol. 2013;10:472–484. doi: 10.1038/nrclinonc.2013.110. [DOI] [PubMed] [Google Scholar]
  • 27.Fortuna G.M.G., Dvir K. Circulating tumor DNA: where are we now? A mini review of the literature. World J Clin Oncol. 2020;11:723–731. doi: 10.5306/wjco.v11.i9.723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kustanovich A., Schwartz R., Peretz T., Grinshpun A. Life and death of circulating cell-free DNA. Cancer Biol Ther. 2019;20:1057–1067. doi: 10.1080/15384047.2019.1598759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Pinzani P., D’Argenio V., Del Re M., Pellegrini C., Cucchiara F., Salvianti F., Galbiati S. Updates on liquid biopsy: current trends and future perspectives for clinical application in solid tumors. Clin Chem Lab Med. 2021;59:1181–1200. doi: 10.1515/cclm-2020-1685. [DOI] [PubMed] [Google Scholar]
  • 30.Schwarzenbach H., Hoon D.S.B., Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011;11:426–437. doi: 10.1038/nrc3066. [DOI] [PubMed] [Google Scholar]
  • 31.Thierry A.R., El Messaoudi S., Gahan P.B., Anker P., Stroun M. Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev. 2016;35:347–376. doi: 10.1007/s10555-016-9629-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wan J.C.M., Massie C., Garcia-Corbacho J., Mouliere F., Brenton J.D., Caldas C., Pacey S., Baird R., Rosenfeld N. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat Rev Cancer. 2017;17:223–238. doi: 10.1038/nrc.2017.7. [DOI] [PubMed] [Google Scholar]
  • 33.Lee J.H., Long G.V., Menzies A.M., Lo S., Guminski A., Whitbourne K., Peranec M., Scolyer R., Kefford R.F., Rizos H., Carlino M.S. Association between circulating tumor DNA and pseudoprogression in patients with metastatic melanoma treated with anti–programmed cell death 1 antibodies. JAMA Oncol. 2018;4:717. doi: 10.1001/jamaoncol.2017.5332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Cullinane C., Fleming C., O’Leary D.P., Hassan F., Kelly L., O’Sullivan M.J., Corrigan M.A., Redmond H.P. Association of circulating tumor DNA with disease-free survival in breast cancer: a systematic review and meta-analysis. JAMA Netw Open. 2020;3 doi: 10.1001/jamanetworkopen.2020.26921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Cabel L., Riva F., Servois V., Livartowski A., Daniel C., Rampanou A., Lantz O., Romano E., Milder M., Buecher B., Piperno-Neumann S., Bernard V., Baulande S., Bieche I., Pierga J.Y., Proudhon C., Bidard F.C. Circulating tumor DNA changes for early monitoring of anti-PD1 immunotherapy: a proof-of-concept study. Ann Oncol. 2017;28:1996–2001. doi: 10.1093/annonc/mdx212. [DOI] [PubMed] [Google Scholar]
  • 36.Creemers A., Krausz S., Strijker M., van der Wel M.J., Soer E.C., Reinten R.J., Besselink M.G., Wilmink J.W., van de Vijver M.J., van Noesel C.J.M., Verheij J., Meijer S.L., Dijk F., Bijlsma M.F., van Oijen M.G.H., van Laarhoven H.W.M. Clinical value of ctDNA in upper-GI cancers: a systematic review and meta-analysis. Biochim Biophys Acta Rev Cancer. 2017;1868:394–403. doi: 10.1016/j.bbcan.2017.08.002. [DOI] [PubMed] [Google Scholar]
  • 37.Guven D.C., Sahin T.K., Yildirim H.C., Aktepe O.H., Dizdar O., Yalcin S. A systematic review and meta-analysis of the association between circulating tumor DNA (ctDNA) and prognosis in pancreatic cancer. Crit Rev Oncol Hematol. 2021;168 doi: 10.1016/j.critrevonc.2021.103528. [DOI] [PubMed] [Google Scholar]
  • 38.Chabon J.J., Simmons A.D., Lovejoy A.F., Esfahani M.S., Newman A.M., Haringsma H.J., Kurtz D.M., Stehr H., Scherer F., Karlovich C.A., Harding T.C., Durkin K.A., Otterson G.A., Purcell W.T., Camidge D.R., Goldman J.W., Sequist L.V., Piotrowska Z., Wakelee H.A., Neal J.W., Alizadeh A.A., Diehn M. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat Commun. 2016;7 doi: 10.1038/ncomms11815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Qian C., Dai N., Xu M., Luo H., Feng Y., Zhang M., Chen R., Wang D. ctDNA facilitated the diagnosis of a patient with synchronous urothelial carcinoma and non-small cell lung cancer: case report. Ann Transl Med. 2020;8:1323. doi: 10.21037/atm-20-6552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Strijker M., Soer E.C., Pastena M., Creemers A., Balduzzi A., Beagan J.J., Busch O.R., Delden O.M., Halfwerk H., Hooft J.E., Lienden K.P., Marchegiani G., Meijer S.L., Noesel C.J., Reinten R.J., Roos E., Schokker S., Verheij J., Vijver M.J., Waasdorp C., Wilmink J.W., Ylstra B., Besselink M.G., Bijlsma M.F., Dijk F., Laarhoven H.W. Circulating tumor DNA quantity is related to tumor volume and both predict survival in metastatic pancreatic ductal adenocarcinoma. Int J Cancer. 2020;146:1445–1456. doi: 10.1002/ijc.32586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ulivi P., Passardi A., Marisi G., Chiadini E., Molinari C., Canale M., Pasini L., Ferroni F., Frassineti G.L., Bartolini G., Monti M. Case report: the added value of liquid biopsy in advanced colorectal cancer from clinical case experiences. Front Pharmacol. 2021;12:745701. doi: 10.3389/fphar.2021.745701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Udomruk S., Orrapin S., Pruksakorn D., Chaiyawat P. Size distribution of cell-free DNA in oncology. Crit Rev Oncol Hematol. 2021;166 doi: 10.1016/j.critrevonc.2021.103455. [DOI] [PubMed] [Google Scholar]
  • 43.Underhill H.R., Kitzman J.O., Hellwig S., Welker N.C., Daza R., Baker D.N., Gligorich K.M., Rostomily R.C., Bronner M.P., Shendure J. Fragment length of circulating tumor DNA. PLoS Genet. 2016;12 doi: 10.1371/journal.pgen.1006162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ungerer V., Bronkhorst A.J., Van den Ackerveken P., Herzog M., Holdenrieder S. Serial profiling of cell-free DNA and nucleosome histone modifications in cell cultures. Sci Rep. 2021;11:9460. doi: 10.1038/s41598-021-88866-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Aucamp J., Bronkhorst A.J., Badenhorst C.P.S., Pretorius P.J. The diverse origins of circulating cell-free DNA in the human body: a critical re-evaluation of the literature: the diverse origins of circulating cell-free DNA. Biol Rev. 2018;93:1649–1683. doi: 10.1111/brv.12413. [DOI] [PubMed] [Google Scholar]
  • 46.Moss J., Magenheim J., Neiman D., Zemmour H., Loyfer N., Korach A., Samet Y., Maoz M., Druid H., Arner P., Fu K.-Y., Kiss E., Spalding K.L., Landesberg G., Zick A., Grinshpun A., Shapiro A.M.J., Grompe M., Wittenberg A.D., Glaser B., Shemer R., Kaplan T., Dor Y. Comprehensive human cell-type methylation atlas reveals origins of circulating cell-free DNA in health and disease. Nat Commun. 2018;9:5068. doi: 10.1038/s41467-018-07466-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Fleischhacker M., Schmidt B. Circulating nucleic acids (CNAs) and cancer--a survey. Biochim Biophys Acta Rev Cancer. 2007;1775:181–232. doi: 10.1016/j.bbcan.2006.10.001. [DOI] [PubMed] [Google Scholar]
  • 48.Holdenrieder S., Von Pawel J., Nagel D., Stieber P. Long-term stability of circulating nucleosomes in serum. Anticancer Res. 2010;30:1613–1615. [PubMed] [Google Scholar]
  • 49.Chen K., Zhao H., Shi Y., Yang F., Wang L.T., Kang G., Nie Y., Wang J. Perioperative dynamic changes in circulating tumor DNA in patients with lung cancer (DYNAMIC) Clin Cancer Res. 2019;25:7058–7067. doi: 10.1158/1078-0432.CCR-19-1213. [DOI] [PubMed] [Google Scholar]
  • 50.Boniface C.T., Spellman P.T. Blood, toil, and taxoteres: biological determinates of treatment-induce ctDNA dynamics for interpreting tumor response. Pathol Oncol Res. 2022;28 doi: 10.3389/pore.2022.1610103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Egger ME, Alexander E, Van Meter T, Kong M-Y, Maung AA, Valdes R Jr, Barousse Hall M, Linder MW Corresponding ctDNA and tumor burden dynamics in metastatic melanoma patients on systemic treatment. Transl Oncol. 2024;42 doi: 10.1016/j.tranon.2024.101883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Cheng F., Su L., Qian C. Circulating tumor DNA: a promising biomarker in the liquid biopsy of cancer. Oncotarget. 2016;7:48832–48841. doi: 10.18632/oncotarget.9453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Avanzini S., Kurtz D.M., Chabon J.J., Moding E.J., Hori S.S., Gambhir S.S., Alizadeh A.A., Diehn M., Reiter J.G. A mathematical model of ctDNA shedding predicts tumor detection size. Sci Adv. 2020;6 doi: 10.1126/sciadv.abc4308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Vioritto I.C.B., Nikolov N., Siegel R.M. Autoimmunity versus tolerance: can dying cells tip the balance? Clin Immunol. 2007;122:125–134. doi: 10.1016/j.clim.2006.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Bettegowda C., Sausen M., Leary R.J., Kinde I., Wang Y., Agrawal N., et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6 doi: 10.1126/scitranslmed.3007094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Del Monte U. Does the cell number 109 still really fit one gram of tumor tissue? Cell Cycle. 2009;8:505–506. doi: 10.4161/cc.8.3.7608. [DOI] [PubMed] [Google Scholar]
  • 57.Sender R., Milo R. The distribution of cellular turnover in the human body. Nat Med. 2021;27:45–48. doi: 10.1038/s41591-020-01182-9. [DOI] [PubMed] [Google Scholar]
  • 58.Franco R.S. Measurement of red cell lifespan and aging. Transfus Med Hemotherapy. 2012;39:302–307. doi: 10.1159/000342232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wood R.L. Regeneration of liver and kidney. JAMA. 1971;218:1306–1307. [Google Scholar]
  • 60.Magrassi L., Leto K., Rossi F. Lifespan of neurons is uncoupled from organismal lifespan. Proc Natl Acad Sci U S A. 2013;110:4374–4379. doi: 10.1073/pnas.1217505110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Green D.R., Evan G.I. A matter of life and death. Cancer Cell. 2002;1:19–30. doi: 10.1016/s1535-6108(02)00024-7. [DOI] [PubMed] [Google Scholar]
  • 62.Labi V., Erlacher M. How cell death shapes cancer. Cell Death Dis. 2015;6:e1675. doi: 10.1038/cddis.2015.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Liu S., Edgerton S.M., Moore D.H., II, Thor A.D. Measures of cell turnover (proliferation and apoptosis) and their association with survival in breast cancer. Clin Cancer Res. 2001;7:1716–1723. [PubMed] [Google Scholar]
  • 64.Kunac N., Sundov Z., Vilovic K. Apoptosis as a prognostic factor in colorectal carcinoma: comparison of TUNEL method and immunohistochemical expression of caspase-3. Appl Immunohistochem Mol Morphol. 2019;27:e22–e27. doi: 10.1097/PAI.0000000000000623. [DOI] [PubMed] [Google Scholar]
  • 65.Heatley M.K. Association between the apoptotic index and established prognostic parameters in endometrial adenocarcinoma. Histopathology. 1995;27:469–472. doi: 10.1111/j.1365-2559.1995.tb00312.x. [DOI] [PubMed] [Google Scholar]
  • 66.Aihara M., Truong L.D., Dunn J.K., Wheeler T.M., Scardino P.T., Thompson T.C. Frequency of apoptotic bodies positively correlates with Gleason grade in prostate cancer. Hum Pathol. 1994;25:797–801. doi: 10.1016/0046-8177(94)90249-6. [DOI] [PubMed] [Google Scholar]
  • 67.Alenzi F.Q.B. Links between apoptosis, proliferation and the cell cycle. Br J Biomed Sci. 2004;61:99–102. doi: 10.1080/09674845.2004.11732652. [DOI] [PubMed] [Google Scholar]
  • 68.Loftus L.V., Amend S.R., Pienta K.J. Interplay between cell death and cell proliferation reveals new strategies for cancer therapy. Int J Mol Sci. 2022;23:4723. doi: 10.3390/ijms23094723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Ryoo H.D., Bergmann A. The role of apoptosis-induced proliferation for regeneration and cancer. Cold Spring Harbor Perspect Biol. 2012;4 doi: 10.1101/cshperspect.a008797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Klemm J., Stinchfield M.J., Harris R.E. Necrosis-induced apoptosis promotes regeneration in drosophila wing imaginal discs. Genetics. 2021;219 doi: 10.1093/genetics/iyab144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Rohne P., Wolf S., Dorr C., Ringen J., Holtz A., Gollan R., Renner B., Prochnow H., Baiersdorfer M., Koch-Brandt C. Exposure of vital cells to necrotic cell lysates induce the IRE1[alpha] branch of the unfolded protein response and cell proliferation. Cell Stress Chaperones. 2018;23:77–88. doi: 10.1007/s12192-017-0825-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Zimmerman M.A., Huang Q., Li F., Liu X., Li C.-Y. Cell death-stimulated cell proliferation: a tissue regeneration mechanism usurped by tumors during radiotherapy. Semin Radiat Oncol. 2013;23:288–295. doi: 10.1016/j.semradonc.2013.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Galluzzi L., Vitale I., Aaronson S.A., Abrams J.M., Adam D., Agostinis P., et al. Molecular mechanisms of cell death: recommendations of the nomenclature committee on cell death 2018. Cell Death Differ. 2018;25:486–541. doi: 10.1038/s41418-017-0012-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Liu Z-g, Jiao D. Necroptosis, tumor necrosis and tumorigenesis. Cell Stress. 2020;4:1–8. doi: 10.15698/cst2020.01.208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Su Z., Yang Z., Xu Y., Chen Y., Yu Q. Apoptosis, autophagy, necroptosis, and cancer metastasis. Mol Cancer. 2015;14:48. doi: 10.1186/s12943-015-0321-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Chaudhuri A.A., Chabon J.J., Lovejoy A.F., Newman A.M., Stehr H., Azad T.D., Khodadoust M.S., Esfahani M.S., Liu C.L., Zhou L., Scherer F., Kurtz D.M., Say C., Carter J.N., Merriott D.J., Dudley J.C., Binkley M.S., Modlin L., Padda S.K., Gensheimer M.F., West R.B., Shrager J.B., Neal J.W., Wakelee H.A., Loo B.W., Alizadeh A.A., Diehn M. Early detection of molecular residual disease in localized lung cancer by circulating tumor DNA profiling. Cancer Discov. 2017;7:1394–1403. doi: 10.1158/2159-8290.CD-17-0716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Tarazona N., Gimeno-Valiente F., Gambardella V., Zuñiga S., Rentero-Garrido P., Huerta M., Rosello S., Martinez-Ciarpaglini C., Carbonell-Asins J.A., Carrasco F., Ferrer-Martínez A., Bruixola G., Fleitas T., Martín J., Tebar-Martínez R., Moro D., Castillo J., Espi A., Roda D., Cervantes A. Targeted next-generation sequencing of circulating-tumor DNA for tracking minimal residual disease in localized colon cancer. Ann Oncol. 2019;30:1804–1812. doi: 10.1093/annonc/mdz390. [DOI] [PubMed] [Google Scholar]
  • 78.Wang Y., Li L., Cohen J.D., Kinde I., Ptak J., Popoli M., Schaefer J., Silliman N., Dobbyn L., Tie J., Gibbs P., Tomasetti C., Kinzler K.W., Papadopoulos N., Vogelstein B., Olsson L. Prognostic potential of circulating tumor DNA measurement in postoperative surveillance of nonmetastatic colorectal cancer. JAMA Oncol. 2019;5:1118–1123. doi: 10.1001/jamaoncol.2019.0512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Egyud M., Tejani M., Pennathur A., Luketich J., Sridhar P., Yamada E., Stahlberg A., Filges S., Krzyzanowski P., Jackson J., Kalatskaya I., Jiao W., Nielsen G., Zhou Z., Litle V., Stein L., Godfrey T. Detection of circulating tumor DNA in plasma: a potential biomarker for esophageal adenocarcinoma. Ann Thorac Surg. 2019;108:343–349. doi: 10.1016/j.athoracsur.2019.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Qiu B., Guo W., Zhang F., Lv F., Ji Y., Peng Y., Chen X., Bao H., Xu Y., Shao Y., Tan F., Xue Q., Gao S., He J. Dynamic recurrence risk and adjuvant chemotherapy benefit prediction by ctDNA in resected NSCLC. Nat Commun. 2021;12:6770. doi: 10.1038/s41467-021-27022-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Sanz-Garcia E., Zhao E., Bratman S.V., Siu L.L. Monitoring and adapting cancer treatment using circulating tumor DNA kinetics: current research, opportunities, and challenges. Sci Adv. 2022;8 doi: 10.1126/sciadv.abi8618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Diehl F., Li M., Dressman D., He Y., Shen D., Szabo S., Diaz L.A., Goodman S.N., David K.A., Juhl H., Kinzler K.W., Vogelstein B. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Med Sci. 2005;102:16368–16373. doi: 10.1073/pnas.0507904102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Diehl F., Schmidt K., Choti M.A., Romans K., Goodman S., Li M., Thornton K., Agrawal N., Sokoll L., Szabo S.A., Kinzler K.W., Vogelstein B., Diaz L.A., Jr. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14:985–990. doi: 10.1038/nm.1789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Kato K., Uchida J., Kukita Y., Kumagai T., Nishino K., Inoue T., Kimura M., Imamura F. Transient appearance of circulating tumor DNA associated with de novo treatment. Sci Rep. 2016;6 doi: 10.1038/srep38639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Lo Y.M., Leung S.F., Chan L.Y., Chan A.T., Lo K.W., Johnson P.J., Huang D.P. Kinetics of plasma Epstein-Barr virus DNA during radiation therapy for nasopharyngeal carcinoma. Cancer Res. 2000;60:2351–2355. [PubMed] [Google Scholar]
  • 86.Tie J., Kinde I., Wang Y., Wong H.L., Roebert J., Christie M., Tacey M., Wong R., Singh M., Karapetis C.S., Desai J., Tran B., Strausberg R.L., Diaz L.A., Papadopoulos N., Kinzler K.W., Vogelstein B., Gibbs P. Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer. Ann Oncol. 2015;26:1715–1722. doi: 10.1093/annonc/mdv177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kruger S., Heinemann V., Ross C., Diehl F., Nagel D., Ormanns S., Liebmann S., Prinz-Bravin I., Westphalen C.B., Haas M., Jung A., Kirchner T., von Bergwelt-Baildon M., Boeck S., Holdenrieder S. Repeated ctDNA measurements represent a novel and promising tool for early response prediction and therapy monitoring in advanced pancreatic cancer. Ann Oncol. 2018;29:2348–2355. doi: 10.1093/annonc/mdy417. [DOI] [PubMed] [Google Scholar]
  • 88.Tamkovich S., Laktionov P. Cell-surface-bound circulating DNA in the blood: biology and clinical application. IUBMB Life. 2019;71:1201–1210. doi: 10.1002/iub.2070. [DOI] [PubMed] [Google Scholar]
  • 89.Bronkhorst A.J., Ungerer V., Holdenrieder S. Early detection of cancer using circulating tumor DNA: biological, physiological and analytical considerations. Crit Rev Clin Lab Sci. 2019;57:253–269. doi: 10.1080/10408363.2019.1700902. [DOI] [PubMed] [Google Scholar]
  • 90.Greytak S.R., Engel K.B., Parpart-Li S., Murtaza M., Bronkhorst A.J., Pertile M.D., Moore H.M. Harmonizing cell-free DNA collection and processing practices through evidence-based guidance. Clin Cancer Res. 2020;26:3104–3109. doi: 10.1158/1078-0432.CCR-19-3015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Bronkhorst A.J., Ungerer V., Holdenrieder S. The emerging role of cell-free DNA as a molecular marker for cancer management. Biomol Detect Quantif. 2019;17 doi: 10.1016/j.bdq.2019.100087. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The Journal of Molecular Diagnostics : JMD are provided here courtesy of American Society for Investigative Pathology

RESOURCES