Skip to main content

Open Access 16.05.2024 | short review

Liquid profiling for patients with advanced cancer is ready for clinical integration

verfasst von: Samantha O. Hasenleithner, Prof. Mag. Dr. Ellen Heitzer

Erschienen in: memo - Magazine of European Medical Oncology

Summary

Molecular profiling of circulating tumor DNA (ctDNA) to guide treatment decisions has found its way into routine management of patients with advanced cancer. This represents a pivotal advancement in precision oncology, offering a non-invasive and fast-tracked method to detecting clinically relevant biomarkers. With the backing of international oncology guidelines, ctDNA analysis is now a standard approach to consider in molecular diagnostics. Despite the promise of ctDNA in refining treatment strategies through the detection of genomic alterations and treatment-relevant biomarkers with high concordance to tissue biopsies, challenges persist. These include the interpretation of discordances due to tumor heterogeneity, sampling biases, and technical limitations, alongside the differentiation of tumor-derived mutations from clonal hematopoiesis. The current consensus supports the utility of comprehensive genomic profiling (CGP) panels for a broad spectrum of actionable targets, while acknowledging the limitations and advocating for a balanced application of “tissue-first” and “plasma-first” approaches tailored to individual patient scenarios. The essential role of molecular tumor boards (MTBs) is in navigating the complexities of ctDNA data interpretation, thereby ensuring the effective incorporation of liquid biopsy into personalized cancer treatment regimens.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

The integration of liquid biopsy into clinical practice marks a significant advancement in the realm of precision oncology, signaling a shift towards more refined and patient-centric approaches in routine cancer care. Clinicians are increasingly performing molecular profiling on circulating tumor DNA (ctDNA) obtained from plasma to non-invasively monitor residual disease [1], guide treatment options [24], or to match patients to suitable clinical trials [5], efforts which are underscored by the issuance of comprehensive implementation guidelines from international oncology and molecular pathology consortia such as ASCO/CAP [6, 7], ESMO [8], NCCN [9, 10], and IASLC. Certain indications now even exist that support the “plasma first” use case, meaning that traditional tissue biopsies may be bypassed in favor of a minimally invasive testing strategy. One example would be the testing for ESR1 mutations in plasma at endocrine resistance in breast cancer to guide addition of selective estrogen receptor degraders. While ctDNA-based assays are redefining clinical pathways, it can be difficult to sift through the growing literature base and clinical trial evidence [11], impeding the adoption of liquid biopsy in everyday cancer management. In this short review, we focus on the advanced cancer setting and offer brief, high-level summaries of the current guidelines, molecular profiling strategies and everyday challenges for incorporating liquid biopsy into real-world precision oncology approaches.

Concordance between alterations in tumor tissue and ctDNA

Many studies have assessed the concordance of genomic alterations detected in major driver genes and treatment-relevant biomarkers between plasma and tissue and have found consistent sensitivities, ranging approximately between 70–90% across various solid tumors [5, 1214]. Discordance is mostly related to tumor heterogeneity and temporal dynamics, sampling biases and technical limitations. Tumors are often heterogeneous and the portion of the tumor sampled for tissue analysis may not fully represent the entire genetic landscape of the tumor, whereas ctDNA represents a mixture of DNA shed from various tumor regions. Moreover, tumor genomes evolve over time due to therapeutic pressure and clonal selection. The ctDNA shed into the bloodstream reflects the most recent state of the tumor, whereas tissue samples may have been obtained at an earlier stage of the disease and may not capture these changes. On the other hand, mutations derived from the hematopoietic system, which can lead to clonal expansions, can be picked up in cfDNA. Clonal hematopoiesis-related mutations can introduce specificity concerns in ctDNA analysis. Without careful validation and discrimination strategies, there is a risk of misinterpreting clonal hematopoiesis-related mutations as tumor-derived mutations, leading to false-positive results.
Taken together, tissue and ctDNA analysis each have strengths and limitations, and their findings may be complementary rather than identical. However, a robust detection agreement among key driver events as well as a comparable number of targetable alterations between ctDNA and tissue profiling [1517] has established the viability of ctDNA-based assays as a substitute for tissue-based testing.

Genomic profiling of ctDNA in patients with advanced cancer for treatment selection

Selecting the right testing approach

Currently, the standard clinical application of genomic profiling of ctDNA is for treatment selection in the advanced cancer setting, meaning detecting alterations that can be matched to targeted therapies or identifying alterations that would be a contraindication for a particular therapy. As with tissue analysis, several scenarios may warrant the limited analysis of a single gene [1820] or using a cancer hotspot panel ([4, 21]; Table 1). However, the general direction of the treatment selection setting is moving toward employing larger comprehensive genomic profiling (CGP) panels to maximize the detection of therapeutic targets and to harvest the additional, valuable information such as mutations, copy numbers, fusions, tumor fraction in plasma (Fig. 1; [22, 23]). In fact, several ctDNA-based CGP tests have already received FDA approval/clearance for select indications (Table 2). Putting algorithmically estimated levels of plasma tumor fraction (TF) into context with the detected variant allele frequencies (VAF) of detected mutations can help with the interpretation of detected mutations and indicate whether they are derived from the germline, from subclones or even from the hematopoietic system (Fig. 2). For CGP panels, TF is usually estimated based on tumor aneuploidy measured as deviations in coverage across the genome. When TFs are below 5–10%, such an estimate is no longer informative, while SNVs can still reliably be detected down to 0.1% [16]. If the panel is large enough, also complex biomarkers such as microsatellite instability (MSI) status and blood tumor mutational burden (bTMB), which have significant relevance for immunotherapies, can be inferred [24]. The added insight that can only be obtained from CGP approaches is critical to downstream interpretation of ctDNA results and provides the best comprehensive overview of the patient’s sample (Table 1).
Table 1
Comparisons and justifications of current molecular testing approaches
Test
Example use case
Rationale for approach
Benefit
Challenges/limitations
Single-gene testing
Determining neoRAS WT
Anti-EGFR rechallenge in mCRC
Quick
Cost-effective
Scalable
Interpretation of a negative result without knowing the tumor content in the sample
Emergence of ESR1 resistance-related mutations in ER+/HER2-negative breast cancer
SERD treatment to counteract endocrine resistance due to ESR1 mutations
Quick
Cost-effective
Scalable
Interpretation of a negative result without knowing the tumor content in the sample
Resistance mutations are often subclonal
Detection of PIK3CA activating mutations in HR+/HER2 advanced or metastatic breast cancer
Identification of HR+/HER2− advanced breast cancer who had received endocrine therapy and who may benefit from alpelisib + fulvestrant
Quick
Cost-effective
Scalable
Interpretation of negative results
Hotspot panels (amplicon)
Lung cancer-specific panel
Availability of many targeted treatments for NSCLC, simultaneous testing of most relevant actionable targets
Quick
Cost-effective
Simple to interpret
Detection of somatic copy number alterations (SCNAs) may not be reliable
Relevant biomarkers such as fusions, MSI, TMB for certain indications are not assessed
Genomic profiling using gene panels
Gene panels (50–150 genes) for treatment selection in patients who have exhausted all standard lines of therapy
Treatment selection in patients who have exhausted all standard lines of therapy
Enables detection of all 4 classes of genomic alterations
Enables an aneuploidy-based estimation of tumor fraction
Higher probability of detecting actionable alterations
Relevant biomarkers such as fusions not included
MSI, TMB cannot be inferred from smaller panels
Variant interpretation is more complex
Distinction of tumor-derived variants, germline variants, and variants derived from clonal hematopoiesis
Often only covers relevant genes for specific tumor entities
CGP panel (> 500 genes) for treatment selection in patients who have exhausted all standard lines of therapy
Treatment selection in patients who have exhausted all standard lines of therapy
Enables detection of all 4 classes of genomic alterations
Enables an aneuploidy-based estimation of the tumor fraction
Pan-cancer suited
Maximization of actionable insight
Inclusion of complex biomarkers like MSI and bTMB
Most expensive
Only cost-effective with a high throughput
Variant interpretation is more complex
Distinction of tumor-derived variants, germline variants, and variants derived from clonal hematopoiesis
High likelihood of detecting multiple co-existing alterations
Requires MTB discussions
Table 2
List of FDA-cleared or FDA-approved companion diagnostic devices (in vitro) for liquid biopsy testing from plasma [32]
Diagnostic Name (Manufacturer)
Indication—Sample Type
Drug Trade Name (Generic) NDA/BLA
Biomarker(s)
Biomarker(s) (Details)
PMA /510(k)/513(f)(2)/HDE (Approval/Clearance/Grant Date)
Agilent Resolution ctDx FIRST assay (Resolution Bioscience, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Plasma
Krazati (adagrasib) NDA 216340
KRAS
KRAS G12C
P210040 (12/12/2022)
Cobas EGFR Mutation Test v2 (Roche Molecular Systems, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Plasma
Tagrisso (osimertinib) NDA 208065
EGFR (HER1)
Exon 19 deletion or exon 21 L858R substitution mutation
P120019/S018 (04/18/2018)
Cobas EGFR Mutation Test v2 (Roche Molecular Systems, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Tissue or Plasma
Iressa (gefitinib) NDA 206995
EGFR (HER1)
Exon 19 deletion or exon 21 L858R substitution mutation
P120019/S019 (08/22/0218)
Cobas EGFR Mutation Test v2 (Roche Molecular Systems, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Tissue or Plasma
Iressa (gefitinib) NDA 206995
EGFR (HER1)
Exon 19 deletion or exon 21 L858R substitution mutation
P120019/S031 (10/27/2020)Group Labeling
Cobas EGFR Mutation Test v2 (Roche Molecular Systems, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Tissue or Plasma
Tarceva (erlotinib) NDA 021743
EGFR (HER1)
Exon 19 deletion or exon 21 L858R substitution mutation
P120019/S031 (10/27/2020)Group Labeling
Cobas EGFR Mutation Test v2 (Roche Molecular Systems, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Tissue or Plasma
Gilotrif (afatinib) NDA 201292
EGFR (HER1)
Exon 19 deletion or exon 21 L858R substitution mutation
P120019/S031 (10/27/2020)Group Labeling
Cobas EGFR Mutation Test v2 (Roche Molecular Systems, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Tissue or Plasma
Tagrisso (osimertinib) NDA 208065
EGFR (HER1)
Exon 19 deletion or exon 21 L858R substitution mutation
P120019/S031 (10/27/2020)Group Labeling
Cobas EGFR Mutation Test v2 (Roche Molecular Systems, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Plasma
Tagrisso (osimertinib) NDA 208065
EGFR (HER1)
T790M
P150044 (09/28/2016)
Cobas EGFR Mutation Test v2 (Roche Molecular Systems, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Plasma
Tarceva (erlotinib) NDA 021743
EGFR (HER1)
Exon 19 deletion or exon 21 L858R substitution mutation
P150047 (06/01/2016)
FoundationOne Liquid CDx (Foundation Medicine, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Plasma
Exkivity (mobocertinib) NDA 215310
EGFR (HER1)
Exon 20 insertion mutations
P190032/S005 (05/03/2023)
FoundationOne Liquid CDx (Foundation Medicine, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Plasma
Iressa (gefitinib) NDA 206995
EGFR (HER1)
Exon 19 deletion or exon 21 L858R substitution mutation
P190032 (08/26/2020)P190032/S008 (12/19/2022)Group Labeling
FoundationOne Liquid CDx (Foundation Medicine, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Plasma
Tagrisso (osimertinib) NDA 208065
EGFR (HER1)
Exon 19 deletion or exon 21 L858R substitution mutation
P190032 (08/26/2020)P190032/S008 (12/19/2022)Group Labeling
FoundationOne Liquid CDx (Foundation Medicine, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Plasma
Tarceva (erlotinib) NDA 021743
EGFR (HER1)
Exon 19 deletion or exon 21 L858R substitution mutation
P190032 (08/26/2020)P190032/S008 (12/19/2022)Group Labeling
FoundationOne Liquid CDx (Foundation Medicine, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Plasma
BRAFTOVI (encorafenib) NDA210496 in combination with MEKTOVI (binimetinib) NDA210498
BRAF
V600E
P190032/S011 (10/11/2023)
FoundationOne Liquid CDx (Foundation Medicine, Inc.)
Metastatic Castrate Resistant Prostate Cancer (mCRPC)—Plasma
Rubraca (rucaparib) NDA 209115
BRCA 1 and BRCA 2
BRCA 1 and BRCA 2 alterations
P190032 (08/26/2020)
FoundationOne Liquid CDx (Foundation Medicine, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Plasma
Tabrecta (capmatinib) NDA 213591
MET
MET single nucleotide variants and indels that lead to MET exon 14 skipping
P190032/S001 (07/15/2021)
FoundationOne Liquid CDx (Foundation Medicine, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Plasma
Rozlytrek (entrectinib) NDA 212725
ROS1
ROS1 fusions
P190032/S004 (12/22/2022)
FoundationOne Liquid CDx (Foundation Medicine, Inc.)
Solid Tumors—Plasma
Rozlytrek (entrectinib) NDA 212725
NTRK1, NTRK2, and NTRK3 fusions
NTRK1/2/3 fusions
P190032/S004 (12/22/2022)
FoundationOne Liquid CDx (Foundation Medicine, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Plasma
Alecensa (alectinib) NDA 208434
ALK
ALK rearrangements
P200006 (10/26/2020)
FoundationOne Liquid CDx (Foundation Medicine, Inc.)
Breast Cancer—Plasma
Piqray (alpelisib) NDA 212526
PIK3CA
C420R, E542K, E545A, E545D [1635G>T only], E545G, E545K, Q546E, Q546R, H1047L, H1047R, and H1047Y
P200006 (10/26/2020)
FoundationOne Liquid CDx (Foundation Medicine, Inc.)
Metastatic Castrate Resistant Prostate Cancer (mCRPC)—Plasma
Lynparza (olaparib) NDA 208558
BRCA 1, BRCA 2 and ATM
BRCA 1, BRCA 2, and ATM alterations
P200006 (10/26/2020)
FoundationOne Liquid CDx (Foundation Medicine, Inc.)
Metastatic Colorectal Cancer (mCRC)—Plasma
BRAFTOVI (encorafenib) NDA 210496 in combination with cetuximab BLA 125084
BRAF
BRAF V600E alteration
P190032/S010 (06/08/2023)
Guardant360 CDx (Guardant Health, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Plasma
Tagrisso (osimertinib) NDA 208065
EGFR (HER1)
EGFR exon 19 deletions, EGFR exon 21 L858R, and T790M
P200010 (08/07/2020)
Guardant360 CDx (Guardant Health, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Plasma
Rybrevant (amivantamb) BLA 761210
EGFR (HER1)
EGFR exon 20 insertions
P200010/S001 (05/21/2021)
Guardant360 CDx (Guardant Health, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Plasma
Lumakras (sotorasib) NDA 214665
KRAS
G12C
P200010/S002 (05/28/2021)
Guardant360 CDx (Guardant Health, Inc.)
Non-Small Cell Lung Cancer (NSCLC)—Plasma
ENHERTU (fam-trastuzumab deruxtecan-nxki) BLA 761139
ERBB2
ERBB2 Activating Mutations (SNVs And Exon 20 Insertions)
P200010/S008 (08/11/2022)
Guardant360 CDx (Guardant Health, Inc.)
Breast Cancer—Plasma
Orserdu (elacestrant) NDA 217639
ESR1
ESR1 missense mutations between codons 310 and 547
P200010/S010 (01/27/2023)
Therascreen PIK3CA RGQ PCR Kit (QIAGEN GmbH)
Breast Cancer—Tissue or Plasma
Piqray (alpelisib) NDA 212526
PIK3CA
C420R, E542K, E545A, E545D [1635G>T only], E545G, E545K, Q546E, Q546R, H1047L, H1047R, and H1047Y
P190001 (05/24/2019)P190004 (05/24/2019)

Selecting the right analyte: tissue first, plasma first, or both in parallel?

An increasing body of evidence supporting the usage, advantages and limitations of plasma-based CGP for guiding treatment decisions has been instrumental in updating clinical guidelines for routine ctDNA testing. This has led to terminologies and concepts such as “tissue-first”, “plasma-first” or matched tissue and liquid profiling, indicating in which clinical scenario it makes sense to perform initial testing on biopsy material, cfDNA from plasma, or both, respectively. Because each approach has its obvious advantages and disadvantages, it is difficult to design a universal testing strategy for patients with advanced cancer and, in the past, recommendations have often conflicted among authors of clinical guidelines [10, 2528] Recently, several of these professional multidisciplinary expert panels have reconvened to provide a general but thorough framework for assay and analyte selection for advanced cancer genotyping which are, for reasons of brevity, summarized here [8, 9]. Generally, it is recognized that ctDNA assays have demonstrated utility in the identification of actionable alterations to inform targeted treatment and may be used routinely for the management of advanced cancer patients, but the assay limitations must be considered. The “tissue-first” approach remains the gold standard for the majority of patients, especially as ctDNA assays are often limited in detecting important events of therapeutic relevance, such as fusions and SCNAs. As such, most guidelines stress the importance of the “tissue-first” approach and generally recommend “plasma-first” for most tumor types when tissue material is unavailable or inadequate [8]. However, a “plasma-first” approach may be performed when a quicker turnaround time is critical for a clinical decision, like in aggressive tumors such as advanced NSCLC [8]. Additionally, there are several scenarios in which ctDNA testing is preferred to standard tissue profiling, such as for the detection of ESR1 mutations in breast cancer and the detection of resistance-related mutations in NSCLC patients who previously received tyrosine kinase inhibitor (TKI) therapy [8]. However, negative ctDNA results can have different interpretations depending on the clinical context and the specific characteristics of the patient and the tumor. While it may suggest the absence of detectable tumor DNA in the bloodstream or a favorable response to treatment, it does not definitively rule out the presence of a mutations, in particular when the sample harbors low tumor content. Therefore, expert guidelines advise reflex tumor testing when non-informative results are obtained. There is also accumulating evidence that performing molecular profiling on both tissue and plasma in parallel significantly enhances the detection of actionable alterations, thus increasing the chance of being able to match patients to targeted therapies [4, 15]. In addition, joint tissue and liquid testing provides valuable complementary, technical and biological information that enables a more holistic interpretation and evaluation of the molecular results.

Interpretation of liquid biopsy data poses challenges for integration into routine clinical care

Much emphasis is put on the challenges of the technical implementation of liquid biopsy in the clinic. However, the complexity of downstream interpretation of molecular testing results from ctDNA is often greatly underestimated (Table 3). The increasing broad coverage and high accuracy of liquid CGP panels has propelled the potential of this technology for guiding treatment decisions, but it comes with everyday challenges in interpreting genomic variance from hundreds of genes and alteration types. This information can only be processed accurately, efficiently and with high confidence within the framework of a clinical team that covers diverse medical disciplines, a construct referred to as the molecular tumor board (MTB) [2931]. MTBs bring together specialists in oncology, pathology, genetics, molecular biology, bioinformatics, patient care and clinical trials. They collaborate to tailor personalized treatment choices for patients, taking into account genomic alterations within their tumors and other relevant clinical factors and data. MTBs may also decide which patients to test, which analytes to assess and which molecular assays to employ. This helps provide clinicians with essential diagnostic, prognostic and actionable insights, enabling them to integrate molecular findings into optimized and individualized care plans for their patients. In routine MTB settings, results from ctDNA testing pose several interpretation challenges that require careful discussion among panel members before reaching treatment or further diagnostic workup recommendations.
Table 3
Select issues and open challenges in interpretation of ctDNA testing results
Issue
Explanation
Solution
Open challenges
When is a liquid biopsy result a true negative? How do you determine this?
Incomplete sensitivity of ctDNA assays poses a risk for false-negative results. In certain scenarios, it may be difficult to differentiate between a non-informative result, i.e. a true negative, or if a variant was undetected because of assay resolution limitations, i.e. false negative
In cases of non-informative results, reflex tissue testing can confirm true negatives. In addition, measuring tumor fraction of the sample is central to determining if sufficient ctDNA levels are present to provide informative results
ESMO guidelines: “Interpretation of a sample as ‘truly negative’ for fusion variants, or copy number variations, using ctDNA remains difficult. Although assays for detection of tumor fraction are in development, they are still experimental, and not available for routine clinical practice.”
Potential germline variants may be detected through liquid CGP. How do you infer potential germline variants and when is there an indication for germline follow-up testing?
Although ctDNA profiling primarily targets somatic mutations, it can also incidentally detect potential germline variants, a factor of which both clinicians and patients should be aware of prior to CGP testing. Detection of potential germline variants necessitates a careful discussion of patient history and a subsequent diagnostic workup
If a variant is present at a high VAF in the absence of extensive tumor shedding in the blood, it may suggest a germline origin. This is particularly relevant when the VAF is around 50%, suggesting that the variant may be present in every cell (as is typical for germline variants).
Some variants detected might be in genes commonly associated with germline mutations. Particular caution must be taken when interpreting pathogenic variants in high penetrance cancer susceptibility genes (such as BRCA 1, BRCA 2, PALB2). If these mutations are known to be common in hereditary cancers and have been documented in germline databases, they might be flagged as potential germline variants. A patient’s personal or family history that suggests a hereditary cancer syndrome is an indication for further germline testing.
Validated germline testing from blood or saliva should be carried out to confirm germline or somatic nature
Before pursuing germline testing, it is essential to obtain informed consent and provide genetic counseling to discuss the implications of the results for the patient and their family. Germline testing raises considerations about privacy, insurance discrimination, and family dynamics, which need careful handling
How do you determine if a variant is CHIP-associated or tumor-specific?
A significant challenge in employing ctDNA-based CGP arises from the absence of standardized approaches for pinpointing the origins of the variants detected in plasma, which includes mutations related to clonal hematopoiesis (CH). Because CH-related variants are not tumor-specific, it is of utmost important to first determine the variant’s origin in order to avoid incorrect treatment matches
Always sequence matched PBMCs at a comparable depth of ctDNA to filter out CH-related mutations
Because an additional sample must be sequenced in parallel to cfDNA, additional costs are incurred, which limits practical application in the clinical setting
Set a threshold, i.e. any variant with a VAF ≥ 0.5% is tumor-specific
The selected threshold may be arbitrary and may still result in the inclusion of CH-related variants or exclusion of tumor-specific variants

Concluding remarks

The accumulating evidence supporting the use of liquid biopsy in routine oncology has paved the way for regulatory approval of several ctDNA-based tests, which has in turn driven an increase in clinical adoption in the advanced disease setting. Currently, one of the main challenges for those starting with liquid biopsy is determining which test best aligns with the clinical question at hand, how many biomarkers should be tested, whether to partner with an academic laboratory or outsource testing to an industry provider, and how to convert NGS readouts into evidence-based treatment decisions, particularly when the results are not as straightforward as with standard tissue testing. While the introduction of CGP has improved the probability of detecting a biomarker-based indication, the additional information provided in liquid biopsy medical reports has posed interpretation challenges that interfere with streamlined treatment decision-making, thus necessitating the interdisciplinary collaboration among medical professionals in the form of an MTB.

Conflict of interest

S.O. Hasenleithner and E. Heitzer declare that they have no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

Neuer Inhalt

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

Literatur
1.
Zurück zum Zitat Semenkovich NP, Szymanski JJ, Earland N, Chauhan PS, Pellini B, Chaudhuri AA. Genomic approaches to cancer and minimal residual disease detection using circulating tumor DNA. J Immunother Cancer. 2023; 01;11(6):e006284. Semenkovich NP, Szymanski JJ, Earland N, Chauhan PS, Pellini B, Chaudhuri AA. Genomic approaches to cancer and minimal residual disease detection using circulating tumor DNA. J Immunother Cancer. 2023; 01;11(6):e006284.
2.
Zurück zum Zitat Brannon RA, Jayakumaran G, Diosdado M, Patel J, Razumova A, Hu Y, et al. Enhanced specificity of clinical high-sensitivity tumor mutation profiling in cell-free DNA via paired normal sequencing using MSK-ACCESS. Nat Commun. 2021; 18;12(1):3770–5.CrossRef Brannon RA, Jayakumaran G, Diosdado M, Patel J, Razumova A, Hu Y, et al. Enhanced specificity of clinical high-sensitivity tumor mutation profiling in cell-free DNA via paired normal sequencing using MSK-ACCESS. Nat Commun. 2021; 18;12(1):3770–5.CrossRef
3.
Zurück zum Zitat Sicklick JK, Kato S, Okamura R, Schwaederle M, Hahn ME, Williams CB, et al. Molecular profiling of cancer patients enables personalized combination therapy: the I‑PREDICT study. Nat Med. 2019; 01;25(5):744–50. CrossRef Sicklick JK, Kato S, Okamura R, Schwaederle M, Hahn ME, Williams CB, et al. Molecular profiling of cancer patients enables personalized combination therapy: the I‑PREDICT study. Nat Med. 2019; 01;25(5):744–50. CrossRef
4.
Zurück zum Zitat Riedl JM, Hasenleithner SO, Pregartner G, Scheipner L, Posch F, Groller K, et al. Profiling of circulating tumor DNA and tumor tissue for treatment selection in patients with advanced and refractory carcinoma: a prospective, two-stage phase II Individualized Cancer Treatment trial. Ther Adv Med Oncol. 2021; 27(13):1758835920987658. Riedl JM, Hasenleithner SO, Pregartner G, Scheipner L, Posch F, Groller K, et al. Profiling of circulating tumor DNA and tumor tissue for treatment selection in patients with advanced and refractory carcinoma: a prospective, two-stage phase II Individualized Cancer Treatment trial. Ther Adv Med Oncol. 2021; 27(13):1758835920987658.
5.
Zurück zum Zitat Rothwell DG, Ayub M, Cook N, Thistlethwaite F, Carter L, Dean E, et al. Utility of ctDNA to support patient selection for early phase clinical trials: the TARGET study. Nat Med. 2019; 01;25(5):738–43.CrossRef Rothwell DG, Ayub M, Cook N, Thistlethwaite F, Carter L, Dean E, et al. Utility of ctDNA to support patient selection for early phase clinical trials: the TARGET study. Nat Med. 2019; 01;25(5):738–43.CrossRef
6.
Zurück zum Zitat Merker JD, Oxnard GR, Compton C, Diehn M, Hurley P, Lazar AJ, et al. Circulating Tumor DNA Analysis in Patients With Cancer: American Society of Clinical Oncology and College of American Pathologists Joint Review. J Clin Oncol. 2018; 01;36(16):1631–41.CrossRef Merker JD, Oxnard GR, Compton C, Diehn M, Hurley P, Lazar AJ, et al. Circulating Tumor DNA Analysis in Patients With Cancer: American Society of Clinical Oncology and College of American Pathologists Joint Review. J Clin Oncol. 2018; 01;36(16):1631–41.CrossRef
7.
Zurück zum Zitat Lockwood CM, Borsu L, Cankovic M, Earle JSL, Gocke CD, Hameed M, et al. Recommendations for Cell-Free DNA Assay Validations: A Joint Consensus Recommendation of the Association for Molecular Pathology and College of American Pathologists. J Mol Diagn. 2023; 01;25(12):876–97. Lockwood CM, Borsu L, Cankovic M, Earle JSL, Gocke CD, Hameed M, et al. Recommendations for Cell-Free DNA Assay Validations: A Joint Consensus Recommendation of the Association for Molecular Pathology and College of American Pathologists. J Mol Diagn. 2023; 01;25(12):876–97.
8.
Zurück zum Zitat Pascual J, Attard G, Bidard F, Curigliano G, De Mattos-Arruda L, Diehn M, et al. ESMO recommendations on the use of circulating tumour DNA assays for patients with cancer: a report from the ESMO Precision Medicine Working Group. Ann Oncol. 2022; 01;33(8):750–68. Pascual J, Attard G, Bidard F, Curigliano G, De Mattos-Arruda L, Diehn M, et al. ESMO recommendations on the use of circulating tumour DNA assays for patients with cancer: a report from the ESMO Precision Medicine Working Group. Ann Oncol. 2022; 01;33(8):750–68.
9.
Zurück zum Zitat Ettinger DS, Wood DE, Aisner DL, Akerley W, Bauman JR, Bharat A, et al. Non-Small Cell Lung Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2022; 01;20(5):497–530. CrossRef Ettinger DS, Wood DE, Aisner DL, Akerley W, Bauman JR, Bharat A, et al. Non-Small Cell Lung Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2022; 01;20(5):497–530. CrossRef
10.
Zurück zum Zitat Gradishar WJ, Anderson BO, Abraham J, Aft R, Agnese D, Allison KH, et al. Breast Cancer, Version 3.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2020; 01;18(4):452–78.CrossRef Gradishar WJ, Anderson BO, Abraham J, Aft R, Agnese D, Allison KH, et al. Breast Cancer, Version 3.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2020; 01;18(4):452–78.CrossRef
11.
Zurück zum Zitat Hasenleithner SO, Speicher MR. A clinician’s handbook for using ctDNA throughout the patient journey. Mol Cancer. 2022; 21;21(1):81–7. CrossRef Hasenleithner SO, Speicher MR. A clinician’s handbook for using ctDNA throughout the patient journey. Mol Cancer. 2022; 21;21(1):81–7. CrossRef
12.
Zurück zum Zitat Adalsteinsson VA, Ha G, Freeman SS, Choudhury AD, Stover DG, Parsons HA, et al. Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat Commun. 2017 06;8(1):1324‑y. Adalsteinsson VA, Ha G, Freeman SS, Choudhury AD, Stover DG, Parsons HA, et al. Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat Commun. 2017 06;8(1):1324‑y.
13.
Zurück zum Zitat Odegaard JI, Vincent JJ, Mortimer S, Vowles JV, Ulrich BC, Banks KC, et al. Validation of a Plasma-Based Comprehensive Cancer Genotyping Assay Utilizing Orthogonal Tissue- and Plasma-Based Methodologies. Clin Cancer Res. 2018 01;24(15):3539–49. Odegaard JI, Vincent JJ, Mortimer S, Vowles JV, Ulrich BC, Banks KC, et al. Validation of a Plasma-Based Comprehensive Cancer Genotyping Assay Utilizing Orthogonal Tissue- and Plasma-Based Methodologies. Clin Cancer Res. 2018 01;24(15):3539–49.
14.
Zurück zum Zitat Park S, Olsen S, Ku BM, Lee M, Jung H, Sun J, et al. High concordance of actionable genomic alterations identified between circulating tumor DNA-based and tissue-based next-generation sequencing testing in advanced non-small cell lung cancer: The Korean Lung Liquid Versus Invasive Biopsy Program. Cancer. 2021 15;127(16):3019–28. Park S, Olsen S, Ku BM, Lee M, Jung H, Sun J, et al. High concordance of actionable genomic alterations identified between circulating tumor DNA-based and tissue-based next-generation sequencing testing in advanced non-small cell lung cancer: The Korean Lung Liquid Versus Invasive Biopsy Program. Cancer. 2021 15;127(16):3019–28.
15.
Zurück zum Zitat Iams WT, Mackay M, Ben-Shachar R, Drews J, Manghnani K, Hockenberry AJ, et al. Concurrent Tissue and Circulating Tumor DNA Molecular Profiling to Detect Guideline-Based Targeted Mutations in a Multicancer Cohort. JAMA Netw Open. 2024 02;7(1):e2351700. Iams WT, Mackay M, Ben-Shachar R, Drews J, Manghnani K, Hockenberry AJ, et al. Concurrent Tissue and Circulating Tumor DNA Molecular Profiling to Detect Guideline-Based Targeted Mutations in a Multicancer Cohort. JAMA Netw Open. 2024 02;7(1):e2351700.
16.
Zurück zum Zitat Husain H, Pavlick DC, Fendler BJ, Madison RW, Decker B, Gjoerup O, et al. Tumor Fraction Correlates With Detection of Actionable Variants Across  23,000 Circulating Tumor DNA. Samples JCO Precis Oncol. 2022;01(6):e2200261.CrossRef Husain H, Pavlick DC, Fendler BJ, Madison RW, Decker B, Gjoerup O, et al. Tumor Fraction Correlates With Detection of Actionable Variants Across  23,000 Circulating Tumor DNA. Samples JCO Precis Oncol. 2022;01(6):e2200261.CrossRef
17.
Zurück zum Zitat Zhang Y, Yao Y, Xu Y, Li L, Gong Y, Zhang K, et al. Pan-cancer circulating tumor DNA detection in over 10,000 Chinese patients. Nat Commun. 2021; 12(1):11–8. Zhang Y, Yao Y, Xu Y, Li L, Gong Y, Zhang K, et al. Pan-cancer circulating tumor DNA detection in over 10,000 Chinese patients. Nat Commun. 2021; 12(1):11–8.
18.
Zurück zum Zitat Osumi H, Takashima A, Ooki A, Yoshinari Y, Wakatsuki T, Hirano H, et al. A multi-institutional observational study evaluating the incidence and the clinicopathological characteristics of NeoRAS wild-type metastatic colorectal cancer. Transl Oncol. 2023; 01(35):101718.CrossRef Osumi H, Takashima A, Ooki A, Yoshinari Y, Wakatsuki T, Hirano H, et al. A multi-institutional observational study evaluating the incidence and the clinicopathological characteristics of NeoRAS wild-type metastatic colorectal cancer. Transl Oncol. 2023; 01(35):101718.CrossRef
19.
Zurück zum Zitat Burstein HJ, DeMichele A, Somerfield MR, Henry NL. Biomarker Testing and Endocrine and Targeted Therapy in Metastatic Breast Cancer Expert Panels. Testing for ESR1 Mutations to Guide Therapy for Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2‑Negative Metastatic Breast Cancer: ASCO Guideline Rapid Recommendation Update. J Clin Oncol. 2023; 20;41(18):3423–5. Jun.CrossRef Burstein HJ, DeMichele A, Somerfield MR, Henry NL. Biomarker Testing and Endocrine and Targeted Therapy in Metastatic Breast Cancer Expert Panels. Testing for ESR1 Mutations to Guide Therapy for Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2‑Negative Metastatic Breast Cancer: ASCO Guideline Rapid Recommendation Update. J Clin Oncol. 2023; 20;41(18):3423–5. Jun.CrossRef
20.
Zurück zum Zitat Suppan C, Graf R, Jahn S, Zhou Q, Klocker EV, Bartsch R, et al. Sensitive and robust liquid biopsy-based detection of PIK3CA mutations in hormone-receptor-positive metastatic breast cancer patients. Br J Cancer. 2022; 126(3):456–63. Suppan C, Graf R, Jahn S, Zhou Q, Klocker EV, Bartsch R, et al. Sensitive and robust liquid biopsy-based detection of PIK3CA mutations in hormone-receptor-positive metastatic breast cancer patients. Br J Cancer. 2022; 126(3):456–63.
21.
Zurück zum Zitat Rothe F, Laes J, Lambrechts D, Smeets D, Vincent D, Maetens M, et al. Plasma circulating tumor DNA as an alternative to metastatic biopsies for mutational analysis in breast cancer. Ann Oncol. 2014; 25(10):1959–65. Rothe F, Laes J, Lambrechts D, Smeets D, Vincent D, Maetens M, et al. Plasma circulating tumor DNA as an alternative to metastatic biopsies for mutational analysis in breast cancer. Ann Oncol. 2014; 25(10):1959–65.
22.
Zurück zum Zitat Zehir A, Benayed R, Shah RH, Syed A, Middha S, Kim HR, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med. 2017; 01;23(6):703–13.CrossRef Zehir A, Benayed R, Shah RH, Syed A, Middha S, Kim HR, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med. 2017; 01;23(6):703–13.CrossRef
23.
Zurück zum Zitat Reitsma M, Fox J, Borre PV, Cavanaugh M, Chudnovsky Y, Erlich RL, et al. Effect of a Collaboration Between a Health Plan, Oncology Practice, and Comprehensive Genomic Profiling Company from the Payer Perspective. J Manag Care Spec Pharm. 2019; 01;25(5):601–11. Reitsma M, Fox J, Borre PV, Cavanaugh M, Chudnovsky Y, Erlich RL, et al. Effect of a Collaboration Between a Health Plan, Oncology Practice, and Comprehensive Genomic Profiling Company from the Payer Perspective. J Manag Care Spec Pharm. 2019; 01;25(5):601–11.
24.
Zurück zum Zitat Kim ES, Velcheti V, Mekhail T, Yun C, Shagan SM, Hu S, et al. Blood-based tumor mutational burden as a biomarker for atezolizumab in non-small cell lung cancer: the phase 2 B‑F1RST trial. Nat Med. 2022; 01;28(5):939–45.CrossRef Kim ES, Velcheti V, Mekhail T, Yun C, Shagan SM, Hu S, et al. Blood-based tumor mutational burden as a biomarker for atezolizumab in non-small cell lung cancer: the phase 2 B‑F1RST trial. Nat Med. 2022; 01;28(5):939–45.CrossRef
25.
Zurück zum Zitat Cardoso F, Paluch-Shimon S, Senkus E, Curigliano G, Aapro MS, Andre F, et al. 5th ESO-ESMO international consensus guidelines for advanced breast cancer (ABC 5). Ann Oncol. 2020; 31(12):1623–49. Cardoso F, Paluch-Shimon S, Senkus E, Curigliano G, Aapro MS, Andre F, et al. 5th ESO-ESMO international consensus guidelines for advanced breast cancer (ABC 5). Ann Oncol. 2020; 31(12):1623–49.
26.
Zurück zum Zitat Planchard D, Popat S, Kerr K, Novello S, Smit EF, Faivre-Finn C, et al. Metastatic non-small cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2018; 29(Suppl 4):iv192–237. Planchard D, Popat S, Kerr K, Novello S, Smit EF, Faivre-Finn C, et al. Metastatic non-small cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2018; 29(Suppl 4):iv192–237.
27.
Zurück zum Zitat Kalemkerian GP, Narula N, Kennedy EB, Biermann WA, Donington J, Leighl NB, et al. Molecular Testing Guideline for the Selection of Patients With Lung Cancer for Treatment With Targeted Tyrosine Kinase Inhibitors: American Society of Clinical Oncology Endorsement of the College of American Pathologists/International Association for the Study of Lung Cancer/Association for Molecular Pathology Clin Practice Guideline Update. J Clin Oncol. 2018; 20;36(9):911–9.CrossRefPubMed Kalemkerian GP, Narula N, Kennedy EB, Biermann WA, Donington J, Leighl NB, et al. Molecular Testing Guideline for the Selection of Patients With Lung Cancer for Treatment With Targeted Tyrosine Kinase Inhibitors: American Society of Clinical Oncology Endorsement of the College of American Pathologists/International Association for the Study of Lung Cancer/Association for Molecular Pathology Clin Practice Guideline Update. J Clin Oncol. 2018; 20;36(9):911–9.CrossRefPubMed
28.
Zurück zum Zitat Burstein HJ, Somerfield MR, Barton DL, Dorris A, Fallowfield LJ, Jain D, et al. Endocrine Treatment and Targeted Therapy for Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2‑Negative Metastatic Breast Cancer: ASCO Guideline Update. J Clin Oncol. 2021; 39(35):3959–77. Burstein HJ, Somerfield MR, Barton DL, Dorris A, Fallowfield LJ, Jain D, et al. Endocrine Treatment and Targeted Therapy for Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2‑Negative Metastatic Breast Cancer: ASCO Guideline Update. J Clin Oncol. 2021; 39(35):3959–77.
29.
Zurück zum Zitat Wahida A, Buschhorn L, Frohling S, Jost PJ, Schneeweiss A, Lichter P, et al. The coming decade in precision oncology: six riddles. Nat Rev Cancer. 2023; 23(1):43–54. Wahida A, Buschhorn L, Frohling S, Jost PJ, Schneeweiss A, Lichter P, et al. The coming decade in precision oncology: six riddles. Nat Rev Cancer. 2023; 23(1):43–54.
30.
Zurück zum Zitat Tsimberidou AM, Kahle M, Vo HH, Baysal MA, Johnson A, Meric-Bernstam F. Molecular tumour boards—current and future considerations for precision oncology. Nat Rev Clin Oncol. 2023; 20(12):843–63. Tsimberidou AM, Kahle M, Vo HH, Baysal MA, Johnson A, Meric-Bernstam F. Molecular tumour boards—current and future considerations for precision oncology. Nat Rev Clin Oncol. 2023; 20(12):843–63.
31.
Zurück zum Zitat Koopman B, Groen HJM, Ligtenberg MJL, Grunberg K, Monkhorst K, de Langen AJ, et al. Multicenter Comparison of Molecular Tumor Boards in The Netherlands: Definition, Composition, Methods, and Targeted Therapy Recommendations. Oncologist. 2021; 26(8):e1347–58. Koopman B, Groen HJM, Ligtenberg MJL, Grunberg K, Monkhorst K, de Langen AJ, et al. Multicenter Comparison of Molecular Tumor Boards in The Netherlands: Definition, Composition, Methods, and Targeted Therapy Recommendations. Oncologist. 2021; 26(8):e1347–58.
Metadaten
Titel
Liquid profiling for patients with advanced cancer is ready for clinical integration
verfasst von
Samantha O. Hasenleithner
Prof. Mag. Dr. Ellen Heitzer
Publikationsdatum
16.05.2024
Verlag
Springer Vienna
Erschienen in
memo - Magazine of European Medical Oncology
Print ISSN: 1865-5041
Elektronische ISSN: 1865-5076
DOI
https://doi.org/10.1007/s12254-024-00978-6

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.