3835 Metabolomics CHAPTER 489
associated with the development or progression of disease. Efforts to
characterize these “metabolic signatures” have been focused primarily
on common, multifactorial diseases such as diabetes, cardiovascular
disease, and various cancers that are well represented in large prospective cohort studies. These studies have, for example, identified altered
levels of amino acids that are associated with a future diagnosis of
diabetes or pancreatic cancer.
Additional efforts have been made to assess the metabolome in
patient samples at the time of an acute presentation. Because altered
metabolite levels can be associated with a specific clinical diagnosis and/
or outcome, the idea is to identify a metabolite signature that facilitates
diagnosis or provides prognostic information. This approach has been
studied, for example, in the context of sepsis and septic shock, in which
blood lactate levels are assessed in combination with the use of clinical
tools such as the Acute Physiology and Chronic Health Evaluation
(APACHE II) or the Sequential Organ Failure Assessment Score (SOFA).
One key limitation in all of these studies is that researchers are primarily assessing correlations between blood plasma metabolite levels
and complex, multisystem diseases. It is often difficult to obtain a
biological understanding of the mechanisms driving these changes or,
even more simply, the primary tissue source(s) of these alterations from
human data alone, without further experimentation in model systems.
■ REFINING DIAGNOSIS AND PREDICTION OF
DRUG SUSCEPTIBILITY
In contrast to the above-described use of metabolomics-based
approaches in multifactorial diseases, the application of these
approaches in some specific contexts can yield an immediate diagnosis and suggest actionable therapeutic interventions. One specific
example in oncology involves an understanding of the pathogenesis of
oncogenic mutations in the metabolic enzyme isocitrate dehydrogenase
(IDH) isoforms 1 and 2. The normal function of these enzymes is to
interconvert isocitrate and α-ketoglutarate; however, cancer-specific
point mutations in these enzymes alter the enzymes’ function in a
manner conferring neomorphic activity that converts isocitrate into
2-hydroxyglutarate (2-HG). 2-HG is a metabolite that is typically
present only at very low levels in cells, but when mutant IDH protein
is present, 2-HG is produced and accumulates to high levels. Elevation
of 2-HG can promote changes that directly contribute to malignancy;
IDH mutations and 2-HG accumulation are found in several human
cancers, including specific clinical subsets of acute myeloid leukemia
and glioma. Given the unique and specific accumulation of 2-HG in
these mutant tumors, detection of this metabolite by LC-MS and NMRbased approaches has been studied both for diagnostic purposes and
as a means of assessing drug response. For example, researchers have
applied MRS-based approaches to assess the accumulation of 2-HG
in gliomas, as this finding can noninvasively identify patients with an
IDH-mutant subset of this cancer (Fig. 489-4). This diagnosis provides
prognostic information and, in the future, may help direct therapeutic options. In principle, metabolomics may identify other disease
biomarkers to aid with diagnosis or therapy assessment in a similar way.
■ PHARMACOMETABOLOMICS
The previous example positions metabolomics as a possible mechanism for achieving a more personalized approach to medicine. The
emerging field of pharmacometabolomics aims to take personalization
further by making this approach more widely applicable across drugs
and disease states. The general workflow is to take a sample population
and perform baseline metabolomics studies on blood from its members. The individuals then receive a given drug with subsequent bloodbased measurement of drug metabolites to gain pharmacokinetics (PK)
and pharmacodynamics (PD) information. These PK and PD data are
then correlated with baseline metabolomic profiling, with the goal of
generating a predictive model for individual PK and PD responses
based on a naïve patient’s metabolomic profile. Correlation with posttreatment metabolomics is also used to provide insight into how
expected responses to therapy could be monitored. Ideally, this
approach would allow clinicians to take a baseline set of measurements
and then—a priori—choose a specific dose of a specific drug to produce
the desired effect in that specific patient. Monitoring of the expected
metabolite changes in response to the drug could also be used to ensure
therapeutic efficacy. If successful, this method could limit both prolonged titration of medications and medication switching, dramatically
shortening and simplifying the current approach to medical therapy.
EMERGING TECHNOLOGIES
While efforts to improve the existing capabilities discussed above are
ongoing, innovations in instrumentation and computation are allowing
collection of metabolite information that previously was not possible.
■ MASS SPECTROMETRY IMAGING
Most clinical metabolomics relies on analysis of bulk material, but
in an individual patient there are areas of normal and diseased tissue, and understanding the differences in metabolism in these areas
requires both spatially sensitive resolution (imaging) and interrogation
(metabolomics). While MRS can perform some of these functions,
it is limited to macroscopic imaging (MRI) and relatively insensitive
metabolomics approaches (NMR). In contrast, MS-based approaches,
while more sensitive, by their nature rely on specimen destruction and
homogenization. The premise of mass spectrometry imaging (MSI)
is to overcome these limitations of MRS and mass spectrometry. MSI
combines histologic evaluation of tissue with MS-based approaches to
assess spatial differences in metabolism. MSI as a technique has been
FIGURE 489-4 In vivo 1
H spectra and analysis demonstrating 2HG detection in IDH mutant brain tumors. A–C In vivo spectra from normal brain (A) and tumors (B–C), are
shown. Components of 2HG, GABA, gluamate and glutamine are displayed. Measurement location indicated by yellow box (voxel). 2HG is seen only in mutant IDH brain
tumors, but not normal brain or wildtype tumors. Shown in brackets is the estimated metabolite concentration (mM) ± s.d. Cho, choline; Cr, creatine; Glu, glutamate; Gln,
glutamine; Gly, glycine; Lac, lactate; Lip, lipids. Scale bars, 1 cm. (Reproduced with permission from Choi et al: 2012).
A B C
3836 PART 20 Frontiers
most highly refined in the neurosciences and can provide subcellular
resolution. In general, thin slices of tissue are mounted on a slide, and
metabolomics is performed at defined points across the slide, yielding
spatial information on where in the tissue section metabolites are measured. One specific approach utilizes matrix-assisted laser desorption/
ionization (MALDI) coupled to MS. In MALDI, tissues are coated
with a special matrix and the MALDI laser scans point-by-point across
a tissue slice, ionizing the metabolites at each location for analysis by
a mass spectrometer. These data can then be referenced back to an
image of the original tissue slice (Fig. 489-5). This approach is being
tested for defining brain tumor margins in real time during resection
and thereby providing insight into boundaries between normal and
abnormal tissues.
■ IMPROVING UNTARGETED METABOLOMICS
Identifying unknown signals in an untargeted metabolomics analysis
remains one of the central challenges in the field. As discussed above,
NMR can definitively identify unknown signals but lags significantly
behind MS-based approaches in its sensitivity and therefore in the
number of signals it can detect. To leverage the sensitivity of MS-based
detection and overcome the challenge of metabolite identification,
researchers are applying computational techniques, using networkstyle analyses to streamline the process. The general approach is to
combine information from known biological perturbations (e.g.,
changes in experimental conditions or disease states), empirical mass
and structural information from MS analysis, and correlations with
known metabolites/pathways to place unknown metabolites within
existing metabolic networks.
SUMMARY
Metabolomics is part of a growing list of “-omics” techniques that have
emerged over recent decades. Despite its immaturity relative to genomics, the power of metabolomics comes from being directly connected to
phenotype and being very sensitive in measuring perturbations within
a system or a patient. While the clinical applications of metabolomics
are currently limited to specific indications, researchers are pushing
to expand these technologies toward broader use in medicine. If these
methods are to be used appropriately, clinicians need to be aware of
established biological and practical confounders. Similarly, a basic
knowledge of the technologies in use and their inherent limitations is
critical. With time and further technical development, metabolomics
could become a routine part of the clinical armamentarium for diagnosis, monitoring, and treatment of disease.
■ FURTHER READING
Bertholdo D et al: Brain proton magnetic resonance spectroscopy:
Introduction and overview. Neuroimaging Clin N Am 23:359, 2013.
Choi C et al: 2-Hydroxyglutarate detection by magnetic resonance
spectroscopy in IDH-mutated patients with gliomas. Nat Med 18:624,
2012.
Emwas AH et al: NMR spectroscopy for metabolomics research.
Metabolites 9:123, 2019.
Gencheva R et al: Clinical benefits of direct-to-definitive testing
for monitoring compliance in pain management. Pain Physician
21:E583, 2018.
Ionization
FIGURE 489-5 Mass spectrometry imaging provides spatial information around metabolites in tissues. Tissue is mounted onto a slide, and a laser or another method is used
to ionize metabolites in a discreet section of the tissue for detection by mass spectrometry. The process is repeated as the laser scans across the tissue, generating an
“image” based on the levels of a metabolite detected at each point in the tissue section.
Kantae V: Integration of pharmacometabolomics with pharmacokinetics and pharmacodynamics: Towards personalized drug therapy.
Metabolomics 13:9, 2017.
Langley RJ et al: An integrated clinico-metabolomic model improves
prediction of death in sepsis. Sci Transl Med 5:195ra95, 2013.
Mayers JR et al: Elevation of circulating branched-chain amino acids
is an early event in human pancreatic adenocarcinoma development.
Nat Med 20:1193, 2014.
Townsend MK et al: Reproducibility of metabolomic profiles among
men and women in 2 large cohort studies. Clin Chem 59:1657, 2013.
Wang TJ et al: Metabolite profiles and the risk of developing diabetes.
Nat Med 17:448, 2011.
While this chapter is focused on the modern use of liquid biopsies and
their applications in the context of malignancies, transplantation, and
noninvasive prenatal testing, it is worth considering that the history
of liquid biopsies dates back many centuries. Indeed, as early as the
fourth century b.c., Hippocrates is noted to have studied body fluids and humors to diagnose various maladies in his patients. By the
mid-seventeenth century, the analysis of bodily fluids (especially urine)
increasingly became a cornerstone of European medicine. However,
arguably, the most important use of bodily fluids as liquid biopsies
currently involves key clinical applications in oncology, solid organ
transplantation, and prenatal diagnosis (Fig. 490-1). Below, we discuss
these applications of liquid biopsies in the context of modern medical
management of patients with risk of cancer and tumor progression,
posttransplantation outcomes, and obstetric management.
CELL-FREE DNA IN ONCOLOGY
Major advances in the twenty-first century in cancer biology have
helped transform oncology, particularly with the advent of personalized cancer therapies, which are typically selected using molecular biomarkers to identify tumor-specific vulnerabilities. However,
because tumor heterogeneity remains a major obstacle to cancer monitoring and treatment, better approaches are needed to define clinically
actionable targets and to facilitate the individualized therapies that
embody precision medicine. While much can be learned from studying tumor biopsies directly, such as molecular profiling that informs
490 Circulating Nucleic Acids
as Liquid Biopsies and
Noninvasive Disease
Biomarkers
Ash A. Alizadeh, Kiran K. Khush,
Yair J. Blumenfeld
3837Circulating Nucleic Acids as Liquid Biopsies and Noninvasive Disease Biomarkers CHAPTER 490
Tumors are well known to release analytes into body
fluids, including the peripheral blood circulation, and the
diagnostic use of such analytes has often been referred to
as liquid biopsies (Fig. 490-2). For hematologic malignancies such as leukemias, the direct examination of
circulating hematopoietic elements is a cornerstone for
diagnosis, response, assessment, and disease monitoring.
Indeed, molecular tools allowed the specific detection of
lymphoma B cells in the blood circulation using in situ
hybridization in the mid-1980s, even before the advent of
polymerase chain reaction for more sensitive detection.
However, even when considering hematologic neoplasms
such as lymphomas, many do not circulate in large numbers in the blood. Indeed, the dominant anatomic distribution of most human cancers results in the absence of
detectable circulating tumor cells as evidence for minimal
residual disease (MRD) in the cellular compartment of
the blood.
Aside from extravasated tumor cells circulating in the
bloodstream as circulating tumor cells (CTCs), liquid
biopsy methods using peripheral blood also include
membrane-bound vesicles released by tumor cells called
exosomes, as well as non–membrane-bound derivatives such as cell-free DNA (cfDNA) and cell-free RNA
(cfRNA), which are released by apoptotic or necrotic
tumor cells. Although the most common source of liquid
biopsy is the peripheral blood, various bodily fluids have
been used for specific liquid biopsy applications to interrogate proximally associated anatomic compartments,
whether using urine, feces, pleural fluid, peritoneal fluid,
bronchoalveolar lavage fluid, saliva, or cerebrospinal
fluid. For example, urinary cfDNA has shown promise for
noninvasive detection of genitourinary tumors including bladder carcinomas, and colorectal cancer screening can be done using multianalyte
assays leveraging fecal DNA.
Among the most intensively studied tumor-derived biomarkers is
circulating tumor DNA in the blood plasma, as a subset of cfDNA.
cfDNA in the blood was first described in 1948 by Mandel and Metais,
• Acute Allograft
Rejection
(dd-cfDNA)
• Microbiome/Virome
• Monitoring Host
Immunity
• Donor-specific
Antibodies
Key Medical
Uses of cfDNA
Liquid Biopsies
Transplant Medicine
• Early Detection
• Noninvasive
Genotyping (ctDNA)
• Molecular Response
Evaluation
• Minimal Residual
Disease (MRD)
Oncology
• Noninvasive
Prenatal Testing
(NIPT/fetal DNA)
• Screening/
Diagnosis
• Fetal Aneuploidies (+21, +18, etc)
• Microdeletions & Duplications
• Single-Gene Disorders
Obstetrics
FIGURE 490-1 Key medical applications of liquid biopsies employing cell-free DNA (cfDNA),
with specific focus on oncology, transplantation, and obstetrics. ctDNA, circulating tumor DNA;
dd-cfDNA, donor-derived cfDNA; NIPT, noninvasive prenatal testing.
diagnosis and monitoring of pathologic response, these are invasive
clinical procedures. In addition, tumor biopsies may not yield enough
material for analysis and can be risky to the patient. Therefore, an
approach to analyze cancer less invasively, such as from a blood sample,
serves as a potentially attractive alternative in the clinical evaluation of
patients with cancer.
Tumor genotyping
MRD detection
Mechanisms of resistance
Circulating
DNA Profiling
Healthy Tissue
Malignant Tissue
FIGURE 490-2 Cell-free DNA (cfDNA) is released both from healthy and malignant tissues. Tumors shed circulating tumor DNA (ctDNA), a minor fraction of all circulating
cfDNA. Molecular profiling of cfDNA enables cancer detection and monitoring applications. MRD, minimal residual disease. (Source: Adapted from BJ Sworder et al:
Hematological Oncology, 39, 2021. https://doi.org/10.1002/hon.6_2879.)
3838 PART 20 Frontiers
Diagnosis
Profiling of tumor DNA
and/or ctDNA using:
Tissue
biopsies
Plasma
samples
Therapy
Subclone 1
Subclone 2
Subclone 3
Surveillance
ctDNA profiling
Tumor evolution
Progression
or disease
transformation
FIGURE 490-3 Framework for noninvasive identification of cancer risk groups.
Schematic illustrating the application of ctDNA profiling for the identification
of adverse risk at different disease milestones. A cancer patient is imagined
as experiencing these disease milestones over time, depicted as an arrow
progressing from left to right. During this temporal sequence, ctDNA can inform
risk at diagnosis, during therapy, in surveillance of disease, and at progression or
disease transformation. At diagnosis, profiling of tumor DNA obtained from either
tissue biopsies (indicated by a scalpel) or plasma (depicted as blood collection
tubes) allows for the identification of patients with high tumor burden and disease
subtypes defined by specific genomic aberrations. Assessment of ctDNA during and
after treatment facilitates the detection of both emerging resistance mutations and
minimal residual disease (MRD) before progression, with potential for noninvasive
prediction of relapse and treatment resistance. Tumor evolution in an anecdotal
patient is illustrated, showing tumor response and clonal evolution over the course
of the disease (detectable subclones at diagnosis are shown in blue/gray; an
emergent subclone after therapy is shown in red). The profiling of tumor DNA and
ctDNA at each milestone is shown by a double-stranded DNA molecule. (Adapted
from F Scherer et al: Distinct biological subtypes and patterns of genome evolution
in lymphoma revealed by circulating tumor DNA. Sci Transl Med 8.364:364ra155-
364ra155, 2016.)
but it was not for another four decades that tumor-derived DNA was
first observed in the plasma of cancer patients. Interestingly, cfDNA is
naturally fragmented with lengths that correlate with DNA wrapped
around individual nucleosomes. Since both healthy cells and cancer
cells release their DNA into the circulation, a major challenge is attaining enough sensitivity to identify and quantify tumor-derived circulating DNA (circulating tumor DNA [ctDNA]) against a potentially large
background of normal constitutional or germline DNA, the majority
of which derives from hematopoietic sources. Methods based on next-generation sequencing offer a way to achieve this. Over the past two
decades, ctDNA has been established as an important biomarker for
studying tumor biology and for detection of cancers. Here, we summarize key applications of ctDNA in the context of early cancer detection,
noninvasive tumor genotyping and classification, molecular response
during therapy, and MRD after definitive therapy (Fig. 490-3).
■ EARLY CANCER DETECTION
For several common cancer types including carcinomas of the lung,
colorectal tract, and breast, early cancer detection via screening can
improve outcomes for adults with established risk factors. However,
such screening can pose significant risks and associated expenses that
limit broad adoption. For example, while annual radiologic screening
by low-dose computed tomography is recommended to screen for lung
cancers in high-risk populations, implementation has been complicated by a high false discovery rate (~90%) and low compliance. Separately, distinct tumor types currently require unique corresponding
screening tests. Finally, most existing cancer screening modalities have
not fully integrated some of the key insights gained through molecular
profiling of cancer genomes. Therefore, there is an unmet need for
new methods for the early detection of cancers. Analysis of ctDNA is
a promising approach that could facilitate blood-based screening. In
this context, several efforts to leverage ctDNA for early detection are
emerging, including approaches focused on individual cancer types,
multianalyte testing combining ctDNA with other biomarkers including proteins, and broader multicancer early detection (MCED) assays
leveraging mutations, tissue-specific methylation, cfDNA fragmentation profiles, and other features.
Despite much excitement about their promise, most such cancer
detection efforts are relatively early in their development. As of 2021,
no screening studies have prospectively applied liquid biopsies as
interventions in randomized studies, with survival outcomes as clinical
endpoints. Indeed, no studies performed to date have demonstrated a
survival advantage of early detection using liquid biopsies in randomized trials, and this remains a key hurdle limiting widespread clinical
adoption of these tests. With these limitations in mind, future prospective studies with substantially longer-term follow-up are needed to
better establish the sensitivity and specificity of MCED tests for early
cancer detection, when considering the common cancer types most
amenable to localized interventions with curative intent. Separately,
because early cancer detection may not improve cancer-specific survival, studies will also be required to determine if these tests can yield
more true positives than false positives, especially because false-positive
results can generate a substantial number of expensive secondary invasive procedures.
■ NONINVASIVE TUMOR GENOTYPING
Molecular subgroups defined by activating mutations in key oncogenic
drivers are the basis for many targeted therapies in oncology across
diverse cancers. For example, targetable mutations in the EGFR gene
are the most common oncogenic driver in non-small-lung cancer
(NSCLC), and the presence of EGFR mutations in NSCLC tumor
biopsies is strongly correlated with a positive response to smallmolecule EGFR tyrosine kinase inhibitors (TKIs). While tissue-based
determination of tumor genotype is still considered by many to represent the gold standard for diagnostic purposes, liquid biopsies have
demonstrated very high positive predictive value for noninvasively
determining tumor genotype in the setting of advanced malignancies.
Indeed, several liquid biopsy assays have been approved by the U.S.
Food and Drug Administration (FDA) for companion diagnostic use
to identify mutations associated with response to targeted therapies in
diverse tumor types. Currently, several comprehensive genomic profiling liquid biopsy assays targeting >50 frequently mutated genes are
currently available as FDA-approved noninvasive tumor genotyping
tests for various solid tumors. For example, liquid biopsy testing using
the Guardant360 test (Guardant Health, Inc.) in patients with NSCLC
can be performed to identify EGFR mutations associated with response
to osimertinib or amivantamab or to identify KRAS mutations associated with sotorasib response. Similarly, liquid biopsy testing using
the FoundationOne Liquid CDx test (Foundation Medicine, Inc.) can
be performed to identify BRCA1/2 gene mutations associated with
rucaparib response in ovarian cancer, ALK rearrangements associated
with alectinib response in NSCLC, PIK3CA mutations associated with
alpelisib response in breast cancer, and BRCA1, BRCA2, and ATM
mutations associated with olaparib response in metastatic castration-resistant prostate cancer.
Of note, such assays have largely been developed for noninvasive
tumor genotyping in the setting of advanced malignancies and are not
well suited in the context of early-stage tumors. Indeed, a recent analysis
by the Sequencing Quality Control Phase 2 (SEQC2) Project Working
Group led by the FDA found that ctDNA detection was less reliable
below a circulating variant allele fraction of 0.5% for mutations of interest. Accordingly, the currently available assays designed for the primary
purpose of noninvasive genotyping in advanced disease are generally
not optimal for detection of MRD because the residual ctDNA allele
fractions after definitive treatment of localized solid cancers are typically much lower than 0.5%. Furthermore, even prior to treatment of
advanced cancers and especially in the context of low metastatic tumor
burden, the modest sensitivity of such liquid biopsy tests currently
requires that if specific lesions of interest are not initially detected in the
blood, a tumor biopsy is still needed to determine if the specific mutations and alterations are present, as this could inform therapy selection.
3839Circulating Nucleic Acids as Liquid Biopsies and Noninvasive Disease Biomarkers CHAPTER 490
■ BIOLOGIC CONSIDERATIONS
In addition to the somatic mutations found in cfDNA that originate
from tumor cells, somatic mutations arising in nontumor tissues can
pose as a source of biologic “background.” Such mutations can confound the use of ctDNA for cancer detection and monitoring. Hematopoietic stem cells can acquire mutations through a process called
age-related clonal hematopoiesis (CH), resulting in variants that can
be found in both cfDNA and circulating peripheral blood leukocytes.
When considering peripheral blood cells in patients who do not otherwise meet criteria for a leukemia diagnosis, identification of mutations
in ~20 genes canonically associated with hematologic neoplasms (and
exceeding 2% in allelic fraction) is termed clonal hematopoiesis of indeterminate potential (CHIP). CHIP has been identified as a risk factor
for cardiovascular disease and hematologic neoplasms and is also
relevant for liquid biopsies (Fig. 490-4). For example, because most
cfDNA derives from hematopoietic sources, CH represents a major
contributor to biologic mutational background for various applications
of liquid biopsies across cancers. Importantly, the prevalence of CH
variants increases with patient age, larger gene panels, and more sensitive testing, with prevalence approaching 100% in adults >60 years old.
Indeed, in liquid biopsy applications for noninvasively identifying
somatic alterations using cfDNA, CH represents the dominant biologic
source of false-positive findings, even if this can vary as a function of
the specific genes or lesions being considered. While CHIP disproportionately affects genes such as DNMT3A, TET2, ASXL1, and JAK2,
which are associated with myeloid cell fitness, population sequencing studies have shown that many more genes in the genome can
be affected by somatic mutations arising during CH in aging adults.
For example, independent liquid biopsy studies have identified that
15–41% of cfDNA mutations in the TP53 gene can be attributable to
CH because these same lesions were found in matched blood leukocytes but not in biopsied tumor tissues.
Accordingly, direct genotyping of peripheral blood leukocytes can
be very helpful to avoid such cfDNA mutations arising from CH,
which can potentially be false-positive results masquerading as ctDNA.
However, studies suggest that ~10% of mutations found in cfDNA of
otherwise healthy adults may not be found in matched leukocytes.
This suggests other noncirculating sources of such cfDNA mutations,
including CH arising in noncirculating hematopoietic precursors
in tissues such as the bone marrow or from clonal proliferations of
nonmalignant, nonhematopoietic cell types. Indeed, recurrent somatic
mutations in genes such as KRAS, MED12, BRAF, and others can
be present in diverse nonmalignant cell types. These mutations (and
others) can arise in a range of epithelial, endothelial, and stromal constituents of vascular malformations, endometrial leiomyomas, melanocytic nevi, and other nonmalignant proliferations. As detailed below,
incorporating sequencing of tumor tissue to initially identify somatic
mutations that are later monitored in plasma samples can help to guard
against these nonmalignant sources of biologic background.
■ PRETREATMENT TUMOR BURDEN
In patients with an established cancer diagnosis, liquid biopsies can be
useful as a noninvasive means of tumor genotyping using plasma, in a
manner that informs therapy selection in such scenarios as described
above. Aside from this use, quantitative assessments of tumor burden
in the plasma prior to therapy can also provide valuable information
via liquid biopsies. For example, pretreatment ctDNA levels have
been shown to have significant associations with established measures
of tumor burden and disease risk including stage, metabolic tumor
volume, and serum protein markers such as lactate dehydrogenase in
lymphomas, CA19-9 levels in pancreatic cancers, and carcinoembryonic antigen levels in colorectal cancers, among others. Importantly,
in many of these scenarios and in other cancers such as NSCLC where
noninvasive tumor biomarkers are not available, pretreatment ctDNA
levels have been shown to have strong prognostic value for treatment
failure and disease progression, providing an independently prognostic
measure of risk. Indeed, such pretreatment ctDNA levels can be used
to noninvasively measure tumor burden poorly captured by other
indices that can lead to biases in clinical trials, including the diagnosis
to treatment interval in lymphomas. Accordingly, pretreatment ctDNA
levels may be used to prevent selection bias in prospective clinical
trials. However, while such independent prognostic value of liquid
biopsies has been validated for several tumors (e.g., using CTC levels
in breast cancers), randomized trials demonstrating the clinical utility
of these measurements for predicting therapeutic benefit from specific
treatments have not yet been performed.
■ MONITORING RESPONSE TO TREATMENT
Beyond the application of liquid biopsies for early cancer detection and
noninvasive tumor genotyping, their use for monitoring therapeutic
responses in a quantitative and qualitative fashion deserves discussion.
In many cancer types, functional imaging has utility for monitoring
systemic response to treatment, when considering relative changes in
volumetric and/or metabolic tumor burden to assess complete versus
partial responses or stable disease. Similarly, in hematologic malignancies such as chronic myelogenous leukemia, the magnitude of response
to systemic therapy with TKIs can be monitored at defined milestones
while on continuous treatment, using defined thresholds for reductions
in BCR-ABL1 transcript levels in the blood. Liquid biopsies can be
similarly useful for measuring quantitative changes in ctDNA while
on therapy. Importantly, as with the use of functional imaging, it is
critical to consider several key factors when using quantitative changes
in ctDNA levels to monitor response. These include the specific tumor
histology and treatment setting (frontline vs relapsed disease), treatment type, the timing of interim ctDNA response assessment, the
type of ctDNA assay being used and associated sensitivity and specificity characteristics, and the thresholds for change in ctDNA levels
observed as informing early molecular response. For example, liquid
biopsy applications for measuring interim responses have been shown
to strongly predict survival outcomes in diffuse large B-cell lymphoma
and Hodgkin’s lymphoma in the frontline setting. Here, 100-fold
reductions (2-log) in ctDNA levels after one cycle of induction chemotherapy have been shown to reliably define an early molecular response
threshold, and 2.5-log reductions in ctDNA levels after two cycles can
be used to define a major molecular response, which are both strongly
associated with event-free and overall survival.
■ MINIMAL RESIDUAL DISEASE
As in the case of several hematologic malignancies, an emerging
body of evidence now demonstrates that liquid biopsies including
ctDNA can detect MRD following treatment of diverse solid tumor
types as a predictor for relapse risk. Indeed, detection of MRD using
ctDNA-based techniques has shown strikingly high positive predictive value for predicting relapse risk in many cancer types, including
carcinomas of the lung, colon, rectum, bladder, and breast, among
others. For example, when considering adjuvant immunotherapy following resection of localized bladder cancers, a retrospective analysis
of a randomized clinical trial strongly suggests that clinical utility for
the checkpoint inhibitor atezolizumab is likely limited only to patients
Nonmalignant
circulating leukocytes
harboring clonal
mutations, called
clonal hematopoiesis
of indeterminate
potential (CHIP), also
release mutant cfDNA
into the blood.
Healthy and cancer
tissues release
cell-free DNA
fragments (cfDNA)
into the blood as
cells turn over.
Tumor
Tissue
Healthy
Tissue
FIGURE 490-4 Contribution of clonal hematopoiesis of indeterminate potential
(CHIP) to cell-free DNA, as relevant for tumor genotyping and monitoring using
circulating tumor DNA. (Source: Adapted from J Boegeholz et al: Hematological
Oncology 39, 2021. https://doi.org/10.1002/hon.23_2879.)
3840 PART 20 Frontiers
with ctDNA MRD detectable in the blood plasma after surgery using
bespoke assays. Separately, the Centers for Medicare and Medicaid
Services recently finalized the first local coverage determination to
provide coverage for ctDNA MRD testing for monitoring colorectal
cancers after surgery using the Signatera MRD test from Natera, and a
broader draft local coverage determination is currently under consideration to enable coverage of ctDNA MRD across tumor types.
Based on these and other similar results, pivotal clinical trials are
now underway that select patients for adjuvant therapy integrating
liquid biopsies to detect MRD and to measure response to such adjuvant and consolidative treatments. Of note, despite the high positive
predictive value of ctDNA MRD for predicting relapse, the clinical
sensitivity of several current assays is generally modest, with a substantial fraction of relapses occurring in patients falsely classified as
MRD negative after initial maneuvers. As in the case of early detection
and noninvasive genotyping described above, for applications of liquid
biopsies to MRD, it is critically important to consider the specific
therapies administered, the biology of ctDNA release, the timing of
MRD measurements, the liquid biopsy assay performance characteristics, and the sources of both technical and biologic background. For
instance, the optimal use of liquid biopsies for their negative predictive
value (e.g., by withholding unnecessary adjuvant therapy in those
without evidence of MRD) is likely to require substantial additional
improvements in the analytical and clinical sensitivity of MRD detection for solid tumors. Nevertheless, although the broad clinical utility
of ctDNA MRD for treatment personalization has yet to be fully established, liquid biopsies hold substantial promise for guiding adjuvant
and consolidative therapies.
■ TECHNICAL CONSIDERATIONS
Despite generally representing a small fraction of nucleic acids that
circulate in the blood plasma, tumor-derived ctDNA molecules can be
identified through a range of techniques related to amplification and
detection. These methods broadly include assays that target tumorspecific mutations, structural variants, somatic copy number alterations, and epigenetic features and generally involve using polymerase
chain reaction (PCR) and high-throughput sequencing. Recently,
substantial improvements in the analytical sensitivity of liquid biopsies
have been achieved through a combination of refinements in these
molecular techniques and the associated computational methods for
analyzing the corresponding data.
Currently, liquid biopsy applications for early cancer detection,
noninvasive tumor genotyping, response monitoring, and MRD rely
mainly either on sequencing-based methods or on amplicon-based
techniques that do not require sequencing, such as digital droplet PCR
(ddPCR) or allele-specific PCR (AS-PCR). Key factors that distinguish
these methods include the cost and turnaround time for the assays
(which are generally in favor of amplicon-based methods), as well as
the scope of genomic aberrations simultaneously being evaluated and
the breadth and depth of the associated molecular profile (which are
generally in favor of sequencing-based methods). For MRD applications, the integration of multiple tumor-specific somatic alterations
into a single assay can allow multiplexed sequencing-based strategies to
achieve analytical sensitivities in the parts-per-million range, especially
in the context of bespoke assays leveraging tumor genotypes.
Among various sequencing-based techniques, liquid biopsy applications have variably profiled the whole human genome or targeted
subregions of the genome, depending on the specific applications
in oncology. More specifically, selective targeting of portions of the
human genome can be achieved either through enrichment by hybridization affinity capture or using locus-specific amplicons, whether
focused on the whole coding exome or smaller, more focused genomic
regions of interest from tens to hundreds of genes.
The biologic and technical sources of background can limit the
sensitivity and specificity of liquid biopsies. Apart from the various
strategies described to reduce these sources of error, many liquid
biopsy studies monitoring therapeutic response and ctDNA MRD
have leveraged tumor genotype–informed analyses to improve performance. Unlike noninvasive genotyping methods that rely entirely on
blood plasma, this tumor genotype–informed approach includes the
profiling of tumor tissue to identify mutations that are then tracked
in posttreatment blood plasma. In reducing the number of mutations
under consideration, this approach reduces the risk of false positives
due to technical and biologic background sources of error. Separately,
the tumor genotype–informed approach can be less demanding for
blood sample volumes. However, due to the very low circulating levels
of tumor-derived DNA at posttreatment milestones, tracking multiple tumor genotype–informed mutations and minimizing unwanted
effects of biologic and technical errors are critical for optimally capturing disease risk in diverse cancers using liquid biopsies to detect
ctDNA MRD.
CELL-FREE DNA IN TRANSPLANTATION
cfDNA testing offers very powerful tools for clinical monitoring of
organ transplant recipients. After transplantation, cfDNA analyses
have been used to assess for development of acute allograft rejection, to
study microbial diversity and infection, and to quantify host immunity.
This section describes current and potential future clinical applications
of cfDNA testing in the transplant arena.
■ NONINVASIVE DETECTION OF ACUTE
ALLOGRAFT REJECTION
In the setting of transplantation, cfDNA is derived from both the recipient tissues and the donated organ or cells. A transplant procedure is
essentially a “genome transplant,” and methods have been developed
to detect and quantify levels of donor-derived cfDNA (dd-cfDNA)
after transplant, with elevated levels reflecting graft injury due to acute
rejection and other forms of graft damage.
In 1998, Lo and colleagues first reported detection of DNA from the
organ donor in the plasma of transplant recipients. By performing PCR
amplification using Y chromosome–specific primers, they were able to
identify dd-cfDNA in the blood of female kidney and liver transplant
recipients. This early work provided proof-of-concept of this unique
approach but was limited to female recipients of male donor organs
(<25% of transplant procedures).
Subsequently, a universal, sex-independent strategy was developed
using whole genome shotgun sequencing to measure single nucleotide
polymorphism (SNP) differences between individuals to quantify the
donor signal. This approach is applicable to any organ donor and recipient combination, regardless of sex, by first genotyping the donor and
recipient to identify sequence differences that can then be used to identify donor cfDNA in the recipient’s blood after transplantation. Prospective studies of this approach have demonstrated that dd-cfDNA is
present at very high levels during the first few days after the transplant
procedure, reflecting cell death within the allograft due to ischemia and
reperfusion injury. Within 1–2 weeks after transplantation, however,
dd-cfDNA levels fall to a low baseline level and remain constant in the
absence of acute rejection.
In the setting of acute rejection, dd-cfDNA levels increase significantly in the transplant recipient’s circulation and correlate with severity of rejection. Initial studies in heart transplantation showed that, at
a threshold value of 0.25%, dd-cfDNA had an area under the receiver
operating characteristic curve (AUC) of 0.60 for distinguishing mild,
0.83 for distinguishing moderate-to-severe, and 0.95 for distinguishing
severe rejection events, each compared to the absence of rejection.
Notably, this assay can be used for surveillance of both acute cellular
and antibody-mediated rejection, as both processes result in graft
damage (Fig. 490-5). A subsequent multicenter study confirmed the
utility of dd-cfDNA monitoring for acute rejection, showing that, at
a threshold of 0.25%, dd-cfDNA had a 99% negative predictive value
for acute rejection and would have safely eliminated 81% of routine
surveillance biopsy procedures.
Notably, increasing dd-cfDNA levels were detected several weeks
to months prior to the rejection event, suggesting that dd-cfDNA is a
highly sensitive marker of graft injury and can enable earlier rejection
diagnosis. Early detection of graft injury may prompt augmentation of
immunosuppressive therapy to halt graft damage in its early stages and
to prevent a subsequent rejection event.
No comments:
Post a Comment
اكتب تعليق حول الموضوع