Next Article in Journal
The Clinical Utility of the NETest in Patients with Small Intestinal Neuroendocrine Neoplasms (Si-NENs): A “Real-Life” Study
Previous Article in Journal
ENO2, a Glycolytic Enzyme, Contributes to Prostate Cancer Metastasis: A Systematic Review of Literature
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Point of Care Liquid Biopsy for Cancer Treatment—Early Experience from a Community Center

1
Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada
2
Division of Advanced Diagnostics, William Osler Health System, Brampton, ON L6R 3J7, Canada
3
Division of Thoracic Surgery, William Osler Health System, Brampton, ON L6R 3J7, Canada
4
Thermo Fisher Scientific, Burlington, ON L7L 5Z1, Canada
5
Division of Medical Oncology, William Osler Health System, Brampton, ON L6R 3J7, Canada
6
Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
7
Division of Medical Oncology, Scarborough Health Network, Scarborough, ON M1P 2V5, Canada
8
Division of Medical Oncology, UHN Princess Margaret Cancer Centre, Toronto, ON M5S 1A1, Canada
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(14), 2505; https://doi.org/10.3390/cancers16142505
Submission received: 11 June 2024 / Revised: 4 July 2024 / Accepted: 8 July 2024 / Published: 10 July 2024
(This article belongs to the Section Methods and Technologies Development)

Abstract

:

Simple Summary

Cancer care is being increasingly driven by biomarker testing. Many biomarkers are traditionally measured on a sample from a patient tumor such as a biopsy. More recently, “liquid biopsy” has emerged as a blood test to complement or replace traditional tissue-based testing. In this report, we explore one of the first case series of rapid liquid biopsy performed within a hospital setting. The results include a median 3-day time to results, compared to approximately 14 days using traditional centralized reference labs. Additional details of the cohort are shared to highlight the utility of point of care liquid biopsy.

Abstract

Liquid biopsy is rapidly becoming an indispensable tool in the oncologist’s arsenal; however, this technique remains elusive in a publicly funded healthcare system, and real-world evidence is needed to demonstrate utility and feasibility. Here, we describe the first experience of an in-house point of care liquid biopsy program at a Canadian community hospital. A retrospective review of consecutive cases that underwent plasma-based next-generation sequencing (NGS) was conducted. Liquid biopsy was initiated at the discretion of clinicians. Sequencing followed a point of care workflow using the Genexus™ integrated sequencer and the Oncomine precision assay, performed by histotechnologists. Results were reported by the attending pathologist. Eligible charts were reviewed for outcomes of interest, including the intent of the liquid biopsy, results of the liquid biopsy, and turnaround time from blood draw to results available. A total of 124 cases, with confirmed or suspected cancer, underwent liquid biopsy between January 2021 and November 2023. The median turnaround time for liquid biopsy results was 3 business days (range 1–12 days). The sensitivity of liquid biopsies was 71%, compared to tissue testing in cases with matched tissue results available for comparison. Common mutations included EGFR (29%), in 86 lung cancer patients, and PIK3CA (22%), identified in 13 breast cancer patients. Healthcare providers ordered liquid biopsies to inform diagnostic investigations and treatment decisions, and to determine progression or resistance mechanisms, as these reasons often overlapped. This study demonstrates that rapid in-house liquid biopsy using point of care methodology is feasible. The technique facilitates precision treatment and offers many additional advantages for cancer care.

1. Introduction

Biomarker-directed therapy represents the current standard of care in the treatment of many types of cancer [1,2]. Providing personalized therapies can improve treatment efficacy and limit side effects often associated with conventional therapies, improving the quality of life and care of cancer patients [2,3,4].
Comprehensive molecular profiling, using next-generation sequencing (NGS), has proven to be valuable in the provision, and development, of targeted therapies. Using multi-gene panels, NGS can identify genomic aberrations and oncogenic drivers in a variety of cancers [1,5,6,7]. In several disease sites, optimal treatments are determined by the sum of multiple different biomarker results [8,9,10,11].
Tissue-based testing is considered the gold standard in biomarker testing practices; however, there are barriers to obtaining, accessing, and utilizing such samples [1,3,6]. Tissue biopsies are invasive, risky, and time-consuming, which can potentially delay treatment. Moreover, the sample itself may not reflect the biology of the tumor due to spatial or temporal heterogeneity [1,2,3].
Plasma-based molecular profiling of circulating tumor molecules (liquid biopsy) presents an opportunity to interrogate tumor-specific biomarkers and inform clinical care without the associated encumbrances of tissue biopsy [7,12]. Liquid biopsy refers to the analysis of circulating biomarkers shed by tumor cells, such as cell-free nucleic acids, circulating tumor cells, proteins, and nucleosomes. These biomarkers are found in bodily fluids including blood, urine, or saliva [1,2,3,7,12]. This paper refers to liquid biopsy as the profiling of circulating tumor DNA/RNA (ctDNA, ctRNA) collected from a peripheral blood draw—a minimally invasive, safe, repeatable, and time-efficient procedure [2,7,13]. These circulating DNA/RNA fragments provide similar information to nucleic acids extracted from a tissue biopsy; with additional relevant information including a quantitative measure of systemic tumor burden [3,12]. Compared to traditional biopsies, liquid biopsy may alleviate the necessity for procurement of tumor tissue, expand access to precision care, and increase accuracy, while decreasing risk to patients and costs to the healthcare system [12,14,15].
Many health systems are faced with challenges in delivering timely and comprehensive biomarker testing. Notable barriers include tissue availability, turnaround time, and cost, among others [2,3]. While liquid biopsy may offer much assistance in overcoming these barriers, the technique is not widely utilized in publicly funded healthcare systems [16]. At the time of this report, liquid biopsy is available through many commercial vendors. The technique is not currently reimbursed in most publicly funded systems, and most hospitals in these regions do not provide in-house liquid biopsy testing. Testing may be accessed only by private-pay, insurance coverage, or other means such as through clinical trials [16]. Despite the promising evidence of liquid biopsy to provide biomarker information at less cost to the healthcare system, its adoption in clinical practice is limited [2,3,7].
Our group has previously implemented rapid in-house or point of care biomarker testing, with an ensuing median turnaround time of 3 business days [17,18]. In parallel to establishing point of care tissue-based testing, our group implemented a rapid in-house point of care liquid biopsy service. In this report, we describe the early clinical experience using point of care liquid biopsy to treat cancer patients in a community setting.

2. Materials and Methods

2.1. Liquid Biopsy Implementation

Liquid biopsy using the Oncomine precision assay and Genexus™ integrated sequencer (Thermofisher, Waltham, MA, USA) was established in January 2021. The assay was validated using a combination of commercial controls, as well as archived plasma samples with matched results from various commercial liquid biopsy assays, showing a high degree of concordance. Following validation, the assay was made available to clinicians at William Osler Health System, Canada, without restriction. Educational sessions were held for medical oncology staff, as well as nursing teams in the oncology inpatient wards and ambulatory clinics. Additional information was provided on an as-needed basis to other specialists including surgeons, respirologists, hospitalists, emergency room physicians, and nurse practitioners. Liquid biopsies were initiated by clinicians for any indication of their choosing. Plasma isolation and sequencing were performed by medical laboratory assistants and immunohistochemistry technologists. All NGS results were interpreted and reported by a molecular pathologist.

2.2. Next-Generation Sequencing of ctDNA/ctRNA

First, 10 mL of peripheral blood was collected in two 5 mL Streck cell-free DNA blood collection tubes. All samples were drawn in-house and received within the lab directly after phlebotomy. No courier services or ambulatory collection was utilized during this period. Upon receipt in the laboratory, the peripheral blood samples were centrifuged at 4000 rpm for 10 min at 4 °C. The plasma portion was aliquoted from the spun sample into DNase/RNase-free Eppendorf microtubes and centrifuged once more at 16,000 rpm for 10 min at 4 °C. The plasma was immediately subjected to nucleic acid extraction, and if not immediately extracted, the supernatant plasma was aliquoted into 2 mL cryogenic tubes and frozen at −80 °C.
Combined cell-free DNA and RNA (cfTNA) extraction was performed with the MagMAX cell-free total nucleic acid isolation kit (Thermofisher, Waltham, MA, USA). Following the manufacturer’s protocol, extraction was performed on plasma from 2 mL or 4 mL volumes depending on the plasma yield. Plasma digestion was performed with proteinase K incubated at 65 °C for 30 min. The digested plasma was then transferred to a processing plate for nucleic acid isolation using the KingFisher Duo Prime Magnetic Particle Processor (Thermofisher, Waltham, MA, USA) resulting in a final eluate volume of 20 μL. The cfTNA was loaded onto the Genexus™ integrated sequencer, and run using the Oncomine precision assay GX, an amplicon-based panel of 50 genes including DNA hotspots and RNA fusions. This assay includes automated library preparation, sequencing, and bioinformatic analysis (Genexus Software 6.2.2, Thermofisher, Waltham, MA, USA) [19].

2.3. Chart Review

To assess the early clinical experience, a retrospective chart review of liquid biopsies performed at William Osler Health System, Canada, between the period January 2021 and November 2023 was conducted. Study approval was provided by the William Osler Health System research ethics board (REB#0118).
Cases were identified using clinical sequencing logs and associated electronic medical records were reviewed. Information collected included demographic and pathology data, as well as the investigative and therapeutic course of the patient’s cancer care journey. An effort to ascertain the underlying reason for the liquid biopsy was made by the study team.
Liquid biopsy reports were reviewed and assessed for concordance where a matched tissue biopsy was available. The determination of whether a tissue biopsy was either matched or concordant was made by the study team upon retrospective review, if not explicitly stated in the liquid biopsy report at the time of interpretation.
Turnaround time was defined as the blood-draw date to the molecular report sign-out date, reported in “business days” by removing weekends and statutory Canadian holidays.

3. Results

From January 2021 to November 2023, a total of 124 consecutive patients underwent liquid biopsy testing. The median age was 67 years (range 23–94 years), with 68 (55%) females. This cohort had predominantly advanced-stage patients, including 3 (2%) with stage III, 114 (92%) with stage IV cancer, and 7 (6%) with unknown staging. The majority of patients (69% (n = 86)) had a lung cancer diagnosis, followed by gastrointestinal malignancies (13% (n = 16)) and breast cancer (10% (n = 13)). Among patients with lung cancer, 55 (64%) of them had no tobacco use history, and 77 (90%) had an adenocarcinoma histologic subtype. Additional demographic data are summarized in Table 1.
From the same time period, same oncology population, and same clinical practitioners, 1759 tissue-based NGS assays were performed in-house.
The median turnaround time for liquid biopsy was 3 business days (interquartile range 2–5 business days). A total of 17 (14%) cases had a turnaround time of 1 day, and 80 cases (65%) were reported in 3 days or less. Turnaround time data are summarized in Figure 1.
Of the 124 sequenced plasma samples, 77 (62%) cases reported alterations and 38% of the cases had no alterations (either negative or nondiagnostic). Among the identified mutations were single nucleotide variants and insertions/deletions (86%), copy number alterations (9%), and fusions (5%). EGFR was the most commonly identified driver mutation in non-small cell lung cancer (29% of cases, n = 86). In gastrointestinal malignancies, KRAS mutation was found in 14% of cases (n = 16), and in breast malignancies, PIK3CA was identified in 22% of cases (n = 13), see Figure 2 and Figure 3.
Overall, 75 cases (61%) had a matched tissue result available for comparison, and a driver alteration was present in 68 (91%) of these cases. Fifty-three (71%) matched tissue and liquid cases were concordant, with 22 cases (29%) demonstrating a driver mutation in a tissue sample only with a false-negative liquid biopsy result. There were no cases (0%) of liquid biopsy with matched tissue biopsy where a driver mutation was found in the liquid biopsy and not in the tissue sample.
Looking separately at alterations identified by ctDNA (SNVs, INDELs, and CNAs), 57 driver alterations were identified, 41 (72%) were detected by both tissue and liquid biopsy, and 16 (28%) were identified by tissue biopsy only. For gene fusions, detected by ctRNA sequencing, 11 cases were identified with matched tissue biopsy. Fusion drivers were detected in both tissue and plasma in five cases (46%), and in a tissue sample only in six cases (55%).
Seven cases showed no driver alteration on tissue testing, all of these cases were driver-negative on liquid biopsy. MET exon 14 skipping alterations could be detected either by ctDNA as an SNV/INDEL or by ctRNA as a MET(13)::MET(15) fusion. Three cases of MET exon 14 skipping were identified with matched tissue samples, the ctDNA alteration was identified in three of three cases (100%), whereas the accompanying ctRNA fusion was identified in one of three cases (33%).
Comparison of liquid biopsy to matched tissue NGS results, where available, are summarized in Table 2.
Actionable driver mutations were identified in eight liquid biopsies (6.5%) where no concurrent tissue sample was available for testing.
As liquid biopsy was available in an unrestricted fashion, user’s intent was extracted from the medical record. The most common reason for performing a liquid biopsy was to guide treatment in 105 cases (85%). Additional uses included: establishing a diagnosis in 32 cases (19%), determining resistance mechanisms to a targeted therapy in 19 cases (11%), as well as determining if a patient’s cancer was progressing on their current non-targeted (chemo- or immunotherapy) therapy in 16 cases (9%). The intent of the liquid biopsy often overlapped, see Figure 4.
In 32 cases where the clinical user was attempting to make a primary cancer diagnosis, 21 (66%) of these liquid biopsies yielded mutational information, and in four cases (13%) a diagnosis was made by liquid biopsy without further invasive tissue sampling.

4. Discussion

Liquid biopsy has emerged as a powerful tool in precision cancer care, offering considerable advantages over, and nicely complementing, tissue-based biomarker testing. This study demonstrates the feasibility and clinical utility of a rapid in-house point of care style technique.
Over an approximately 3-year period, 124 liquid biopsies were reported with a median turnaround time of 3 days. This result is in keeping with previous reports using similar methods [20]. To our knowledge, at the time of this writing, this represents the fastest turnaround time reported for comprehensive liquid NGS. With this study, adding to previous reports, a rapid in-house approach offers a considerable time advantage over the use of commercial labs [21,22,23].
The timeliness of treatment is critical in diseases such as non-small cell lung cancer, where the mortality of untreated advanced disease is 4% per week, and the cost to the healthcare system in providing supportive care is over $400 dollars per week [24,25]. Treatment initiation without biomarker data can have a severely adverse effect on patient outcomes [26]. The median 3-day turnaround time reported here for liquid biopsy, and previously for tissue biopsy, represents practice-changing advances in the delivery of biomarkers to support precision cancer care as a standard practice in community settings [17]. Variability in the turnaround time (range 1–12 days) was due to technologist and sequencing instrument availability. In-house biomarker testing is prioritized according to patient needs, with faster turnaround times provided when indicated. In this series, 14% of liquid biopsies were reported within 24 h of blood draw to accommodate stat and urgent requests.
Negative liquid biopsies found no results. Non-diagnostic liquid biopsies failed to detect a known driver mutation or failed to meet appropriate quality metrics. It is often difficult to distinguish a non-diagnostic sample from a negative sample, especially in scenarios with no prior biomarker information available. As liquid biopsy uptake continues, clinician users may benefit from support around the interpretation and appropriate response to results, particularly around negative/nondiagnostic tests.
The overall concordance of liquid biopsy to matched tissue results was relatively high; however, this was driven largely by point mutations and other alterations identified in ctDNA. The liquid biopsy assay utilizes ctRNA to identify fusions. This performed relatively poorly with an overall sensitivity of only 46% compared to matched tissue results. The results suggest that this is not a reliable method for the detection of oncogenic fusions in plasma. The phenomenon is nicely demonstrated by MET exon 14 skipping alterations which can be detected in both the ctDNA and ctRNA components of the panel. Here we reported three patients, identified by matched tissue biopsy, to harbor a MET exon 14 skipping driver mutation. All three were detected in the form of a ctDNA splice site alteration; however, only one of the three was concurrently detected in the ctRNA space as a MET(13)::MET(15) fusion. Typically, tissue-based NGS shows the opposite pattern, with RNA sequencing demonstrating a higher sensitivity [27].
The majority of commercially available liquid biopsy assays utilize a ctDNA-only approach. In the plasma space, reports from ctDNA hybrid capture-based approaches have shown superior sensitivity for the detection of ALK fusions [28]. Previous studies have shown the Oncomine precision assay to have a high sensitivity for fusions, particularly at low RNA concentrations [29]. In the setting of liquid biopsy, the paradoxically low sensitivity for fusions is believed to be pre-analytic in nature.
Future improvements to blood collection and transport, as well as ctRNA extraction from plasma, may improve upon the sensitivity. While Streck blood collection tubes and transport of peripheral blood in ambient temperature do not impact ctDNA, it is likely to affect ctRNA viability [30]. As such, blood collection tubes that can also preserve ctRNA and temperature-controlled transport of peripheral blood may be beneficial; however, further research is required. Until that time, users should be aware of this limitation, particularly in diseases where oncogenic fusions are relevant to diagnosis and treatment. The low sensitivity for fusions seen in this approach may be balanced against the ease of use, implementation, and rapid turnaround time associated with this methodology.
This study was conducted in a public healthcare setting, where cost remains front of mind, and the price for liquid biopsy is a prominent disincentive for payers. It should be viewed as a very positive signal that over an approximate 3-year study period, 124 liquid biopsy tests were ordered—none were rejected, and no limitations were placed on usage. For comparison, over this same study period, 1759 tissue-based NGS were performed for the same oncology practice group, with liquid biopsy representing less than 7% of all NGS usage. Within many healthcare systems, faster results are often quoted as an underpinning reason for the use of liquid biopsy [2,3]. Within our center, during this study period, rapid tissue-based NGS was also available for patients and providers, and hence within the usage data, liquid biopsy for “faster results” was not observed. In our center, a setting with rapidly-available tissue results, liquid biopsy was requested fewer than 50 times per year or less than once per week. This would represent a limitation of this study as results may not be transferrable to community centers with protracted wait times for biomarkers, or even those with standard 14-day wait times.
Within the usage data, there were two usages identified, which have not previously been well described for liquid biopsy in cancer care. In this series, users attempted to make a primary cancer diagnosis in 32 liquid biopsies. All of these instances would represent a patient with suspected cancer, in whom a tissue diagnosis was either not possible, challenging, or too time-consuming. Additionally, many liquid biopsies were ordered to diagnose progression (outside of acquired resistance to a targeted therapy). In these cases, clinicians would use liquid biopsy data to distinguish cancer progression from mimics such as radiation pneumonitis, infection, and others. These utilizations of liquid biopsy are certainly facilitated by the rapid turnaround time associated with the technique and may not be possible with a turnaround time of 2 weeks or greater. As discussed above, the integration of other available data, including imaging, patient history, and multidisciplinary tumor board discussion is invaluable to the interpretation of liquid biopsy when looking beyond the elucidation of matched targeted therapies.
In this cohort, 21 patients were diagnosed with cancer, and some may have avoided an invasive sampling procedure, associated complications, and tissue-based testing costs and delays. In four cases, a diagnosis was made without a tissue biopsy. The systemic cost savings of in-house liquid biopsy implementation is felt to be large, but further studies are needed to quantify this. The utility in this regard will also likely improve with both clinician and laboratorian experience. While some patients were able to avoid invasive procedures, the utility of tissue biopsy including morphology, histology, and immunohistochemistry remains paramount in precision cancer care.
Many studies have demonstrated the complementary nature of tissue and liquid biopsies, showing that while many actionable genomic alterations are shared between tissue and liquid testing, many are private to either tissue-only or liquid-only [31,32,33,34]. In this cohort, there were no instances where liquid biopsy identified a mutation that was not detected by matched tissue sequencing; however, alterations were identified by plasma-only where a tissue biopsy was unavailable. This dataset is intended to highlight the value of concurrently available rapid point of care liquid and tissue-based NGS in a single center.

5. Conclusions

Rapid in-house liquid biopsy is feasible and can be implemented in community practice to support cancer care. This point of care technique enables a median turnaround time of 3 business days. The technique offers considerable advantages over tissue biopsy alone, but may be associated with lower sensitivity, especially for gene fusions. This study highlights additional clinical utility beyond targeted therapy selection, such as primary cancer diagnosis.
Liquid biopsy is proven to be a valuable test in cancer care. A rapid in-house approach offers considerable advantages over more traditional send-out methods. More work is needed to improve the sensitivity of ctRNA-based NGS to detect oncogenic fusions. Additional studies are needed to measure the economic value and cost effectiveness of the technique.

Author Contributions

Conceptualization, B.S.S., A.B. and P.K.C.; data curation, C.N. and B.S.S.; formal analysis, C.N., B.S.S. and X.W.; funding acquisition, B.S.S.; investigation, A.M.B., S.D., M.I., K.I., K.P., W.R., R.S., D.C.C.T., C.Y. and P.K.C.; methodology, A.B.; protocol development, A.B., K.D. and B.S.S.; project administration, C.N. and B.S.S.; supervision, B.S.S., X.W. and P.K.C.; validation, A.B. and B.S.S.; visualization, C.N. and B.S.S.; writing—original draft preparation, B.S.S., C.N. and X.W.; writing—review and editing, B.S.S., C.N., A.B., A.M.B., K.R.B., S.D., M.I., K.P., W.R., D.C.C.T., X.W. and P.K.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by multiple sources, including research grants from Eli Lilly, Roche (grant number 20040020), Pfizer (grant number 68023243), and ThermoFisher. The work is original in nature and funders had no role in the conceptualization, design, analysis, or writing of this manuscript.

Institutional Review Board Statement

This study was approved by the research ethics board of William Osler Health System in Brampton, ON, Canada (protocol #0118; 19 December 2023).

Informed Consent Statement

Patient consent was waived due to the retrospective nature of this study and minimal risk to patients.

Data Availability Statement

Supporting data can be available from the corresponding author, B.S.S., upon reasonable request.

Acknowledgments

The authors would like to acknowledge the William Osler Health System’s Advanced Diagnostics team.

Conflicts of Interest

K.D. is an employee of ThermoFisher at the time of publication. S.D. has participated in advisory board meetings with Bristol-Myers Squibb, Eisai, Ipsen, Merck, Pfizer, and Taiho and has received honoraria from AstraZeneca, Ipsen, Merck, and Pfizer. M.I. has participated in advisory board meetings with Novartis; clinical trials with Astellas Pharma Global Development Inc. (Northbrook, IL, USA), AstraZeneca, and Bristol-Myers Squibb; and has received honoraria from Bayer, Canadian Urologic Association, CompassMD, Ipsen, MD Analytics, Merck, Save Your Skin, and Sermo Team. K.P. has received continuing medical education speaker honorarium from Merck, and AstraZeneca; and consulting fees from Amgen, Takeda, Merck, Knight pharmaceuticals, AstraZeneca and BMS. W.R. has received honoraria and/or participated in advisory board meetings with Ipsen, Gilead, Knight Therapeutics, Novartis, AstraZeneca, Pfizer, and Merck; and participated in clinical trials with Pfizer and Roche. P.K.C. has received honoraria and/or has participated in advisory board meetings with Amgen, AstraZeneca, Bristol Myers Squibb, EMD Serono, Merck, Janssen, Bayer, Novartis, Pfizer, Roche, and Takeda. B.S.S. has received honoraria, grant support, and/or participated in advisory meetings with Amgen, AstraZeneca, Bayer, Biocartis, Boehringer-Ingelheim, Cell Marque, Elevation Oncology, Eli Lilly, EMD Serono, Incyte, Janssen, Merck, Novartis, Pfizer, Roche, Sanofi, ThermoFisher, and Turning Point Therapeutics. The remaining authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Haselmann, V.; Hedtke, M.; Neumaier, M. Liquid Profiling for Cancer Patient Stratification in Precision Medicine—Current Status and Challenges for Successful Implementation in Standard Care. Diagnostics 2022, 12, 748. [Google Scholar] [CrossRef]
  2. Hirahata, T.; Ul Quraish, R.; Quraish, A.U.; Ul Quraish, S.; Naz, M.; Razzaq, M.A. Liquid Biopsy: A Distinctive Approach to the Diagnosis and Prognosis of Cancer. Cancer Inform. 2022, 21, 117693512210760. [Google Scholar] [CrossRef] [PubMed]
  3. Mathai, R.; Vidya, R.; Reddy, B.; Thomas, L.; Udupa, K.; Kolesar, J.; Rao, M. Potential Utility of Liquid Biopsy as a Diagnostic and Prognostic Tool for the Assessment of Solid Tumors: Implications in the Precision Oncology. J. Clin. Med. 2019, 8, 373. [Google Scholar] [CrossRef] [PubMed]
  4. Hoeben, A.; Joosten, E.A.J.; Van Den Beuken-van Everdingen, M.H.J. Personalized Medicine: Recent Progress in Cancer Therapy. Cancers 2021, 13, 242. [Google Scholar] [CrossRef]
  5. Karlovich, C.A.; Williams, P.M. Clinical Applications of Next-Generation Sequencing in Precision Oncology. Cancer J. 2019, 25, 264–271. [Google Scholar] [CrossRef]
  6. García-Pardo, M.; Leighl, N. “Plasma-First” Approach for Molecular Genotyping in Non-Small Cell Lung Cancer: A Narrative Review. J. Liq. Biopsy 2023, 2, 100123. [Google Scholar] [CrossRef]
  7. Lin, C.; Liu, X.; Zheng, B.; Ke, R.; Tzeng, C.-M. Liquid Biopsy, ctDNA Diagnosis through NGS. Life 2021, 11, 890. [Google Scholar] [CrossRef]
  8. National Comprehensive Cancer Network. Available online: https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf (accessed on 8 March 2024).
  9. National Comprehensive Cancer Network. Available online: https://www.nccn.org/professionals/physician_gls/pdf/colon.pdf (accessed on 8 March 2024).
  10. National Comprehensive Cancer Network. Available online: https://www.nccn.org/professionals/physician_gls/pdf/breast.pdf (accessed on 8 March 2024).
  11. Pascual, J.; Attard, G.; Bidard, F.-C.; Curigliano, G.; De Mattos-Arruda, L.; Diehn, M.; Italiano, A.; Lindberg, J.; Merker, J.D.; Montagut, C.; 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, 33, 750–768. [Google Scholar] [CrossRef] [PubMed]
  12. Cha, Y.; Kim, S.; Han, S.-W. Utilizing Plasma Circulating Tumor DNA Sequencing for Precision Medicine in the Management of Solid Cancers. Cancer Res. Treat. 2023, 55, 367–384. [Google Scholar] [CrossRef]
  13. Serratì, S.; De Summa, S.; Pilato, B.; Petriella, D.; Lacalamita, R.; Tommasi, S.; Pinto, R. Next-Generation Sequencing: Advances and Applications in Cancer Diagnosis. OncoTargets Ther. 2016, 9, 7355–7365. [Google Scholar] [CrossRef]
  14. Ezeife, D.A.; Spackman, E.; Juergens, R.A.; Laskin, J.J.; Agulnik, J.S.; Hao, D.; Laurie, S.A.; Law, J.H.; Le, L.W.; Kiedrowski, L.A.; et al. The Economic Value of Liquid Biopsy for Genomic Profiling in Advanced Non-Small Cell Lung Cancer. Ther. Adv. Med. Oncol. 2022, 14, 175883592211126. [Google Scholar] [CrossRef]
  15. Fagery, M.; Khorshidi, H.A.; Wong, S.Q.; Vu, M.; IJzerman, M. Health Economic Evidence and Modeling Challenges for Liquid Biopsy Assays in Cancer Management: A Systematic Literature Review. PharmacoEconomics 2023, 41, 1229–1248. [Google Scholar] [CrossRef] [PubMed]
  16. Febbo, P.G.; Allo, M.; Alme, E.B.; Cuyun Carter, G.; Dumanois, R.; Essig, A.; Kiernan, E.; Kubler, C.B.; Martin, N.; Popescu, M.C.; et al. Recommendations for the Equitable and Widespread Implementation of Liquid Biopsy for Cancer Care. JCO Precis. Oncol. 2024, 8, e2300382. [Google Scholar] [CrossRef] [PubMed]
  17. Sheffield, B.S.; Beharry, A.; Diep, J.; Perdrizet, K.; Iafolla, M.A.J.; Raskin, W.; Dudani, S.; Brett, M.A.; Starova, B.; Olsen, B.; et al. Point of Care Molecular Testing: Community-Based Rapid Next-Generation Sequencing to Support Cancer Care. Curr. Oncol. 2022, 29, 1326–1334. [Google Scholar] [CrossRef] [PubMed]
  18. Wang, X.; Beharry, A.; Sheffield, B.S.; Cheema, P.K. Feasibility of Point-of-Care Genomic Profiling in the Diagnosis and Treatment of Cancer of Unknown Primary. Oncologist 2023, 28, 474–478. [Google Scholar] [CrossRef] [PubMed]
  19. ThermoFisher Scientific. Available online: https://www.thermofisher.com/ca/en/home/clinical/preclinical-companion-diagnostic-development/oncomine-oncology/oncomine-precision-assay.html (accessed on 12 March 2024).
  20. Low, S.-K.; Ariyasu, R.; Uchibori, K.; Hayashi, R.; Chan, H.T.; Chin, Y.M.; Akita, T.; Harutani, Y.; Kiritani, A.; Tsugitomi, R.; et al. Rapid Genomic Profiling of Circulating Tumor DNA in Non-Small Cell Lung Cancer Using Oncomine Precision Assay with GenexusTM Integrated Sequencer. Transl. Lung Cancer Res. 2022, 11, 711–721. [Google Scholar] [CrossRef] [PubMed]
  21. Guardant. Available online: https://guardanthealth.com/products/tests-for-patients-with-early-and-advanced-stage-cancer/ (accessed on 12 March 2024).
  22. Tempus. Available online: https://www.tempus.com/resources/faqs%20-%20oncology/#:~:text=Tempus%20results%20can%20be%20expected%20~7%E2%80%9310%20da%20ys%20after,not%20begin%20until%20all%20required%20specimens%20are%20received (accessed on 12 March 2024).
  23. Sehayek, O.; Kian, W.; Onn, A.; Stoff, R.; Sorotsky, H.G.; Zemel, M.; Bar, J.; Dudnik, Y.; Nechushtan, H.; Rottenberg, Y.; et al. Liquid First Is “Solid” in Naïve Non-Small Cell Lung Cancer Patients: Faster Turnaround Time With High Concordance to Solid Next-Generation Sequencing. Front. Oncol. 2022, 12, 912801. [Google Scholar] [CrossRef] [PubMed]
  24. Stewart, D.J.; Maziak, D.E.; Moore, S.M.; Brule, S.Y.; Gomes, M.; Sekhon, H.; Dennie, C.; Lo, B.; Fung-Kee-Fung, M.; Bradford, J.; et al. The Need for Speed in Advanced Non-small Cell Lung Cancer: A Population Kinetics Assessment. Cancer Med. 2021, 10, 9040–9046. [Google Scholar] [CrossRef] [PubMed]
  25. Sheffield, B.S.; Eaton, K.; Emond, B.; Lafeuille, M.-H.; Hilts, A.; Lefebvre, P.; Morrison, L.; Stevens, A.L.; Ewara, E.M.; Cheema, P. Cost Savings of Expedited Care with Upfront Next-Generation Sequencing Testing versus Single-Gene Testing among Patients with Metastatic Non-Small Cell Lung Cancer Based on Current Canadian Practices. Curr. Oncol. 2023, 30, 2348–2365. [Google Scholar] [CrossRef]
  26. Aggarwal, C.; Marmarelis, M.E.; Hwang, W.-T.; Scholes, D.G.; McWilliams, T.L.; Singh, A.P.; Sun, L.; Kosteva, J.; Costello, M.R.; Cohen, R.B.; et al. Association Between Availability of Molecular Genotyping Results and Overall Survival in Patients with Advanced Nonsquamous Non–Small-Cell Lung Cancer. JCO Precis. Oncol. 2023, 7, e2300191. [Google Scholar] [CrossRef]
  27. Davies, K.D.; Lomboy, A.; Lawrence, C.A.; Yourshaw, M.; Bocsi, G.T.; Camidge, D.R.; Aisner, D.L. DNA-Based versus RNA-Based Detection of MET Exon 14 Skipping Events in Lung Cancer. J. Thorac. Oncol. 2019, 14, 737–741. [Google Scholar] [CrossRef] [PubMed]
  28. Dziadziuszko, R.; Mok, T.; Peters, S.; Han, J.-Y.; Alatorre-Alexander, J.; Leighl, N.; Sriuranpong, V.; Pérol, M.; De Castro Junior, G.; Nadal, E.; et al. Blood First Assay Screening Trial (BFAST) in Treatment-Naive Advanced or Metastatic NSCLC: Initial Results of the Phase 2 ALK-Positive Cohort. J. Thorac. Oncol. 2021, 16, 2040–2050. [Google Scholar] [CrossRef] [PubMed]
  29. Bormann Chung, C.; Lee, J.; Barritault, M.; Bringuier, P.-P.; Xu, Z.; Huang, W.-Y.; Beharry, A.; Castillo, J.; Christiansen, J.; Lin, J.C.; et al. Evaluating Targeted Next-Generation Sequencing Assays and Reference Materials for NTRK Fusion Detection. J. Mol. Diagn. 2022, 24, 18–32. [Google Scholar] [CrossRef] [PubMed]
  30. Sorber, L.; Zwaenepoel, K.; Jacobs, J.; De Winne, K.; Goethals, S.; Reclusa, P.; Van Casteren, K.; Augustus, E.; Lardon, F.; Roeyen, G.; et al. Circulating Cell-Free DNA and RNA Analysis as Liquid Biopsy: Optimal Centrifugation Protocol. Cancers 2019, 11, 458. [Google Scholar] [CrossRef] [PubMed]
  31. Pritchett, M.A.; Camidge, D.R.; Patel, M.; Khatri, J.; Boniol, S.; Friedman, E.K.; Khomani, A.; Dalia, S.; Baker-Neblett, K.; Plagnol, V.; et al. Prospective Clinical Validation of the InVisionFirst-Lung Circulating Tumor DNA Assay for Molecular Profiling of Patients with Advanced Nonsquamous Non–Small-Cell Lung Cancer. JCO Precis. Oncol. 2019, 3, PO.18.00299. [Google Scholar] [CrossRef] [PubMed]
  32. Park, S.; Olsen, S.; Ku, B.M.; Lee, M.; Jung, H.; Sun, J.; Lee, S.; Ahn, J.S.; Park, K.; Choi, Y.; 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, 127, 3019–3028. [Google Scholar] [CrossRef] [PubMed]
  33. Hao, D.; Laskin, J.; Laurie, S.; Agulnik, J.; Juergens, R.; Ezeife, D.; Law, J.; Le, L.; Kiedrowski, L.; Melosky, B.; et al. P89.03 Demonstrating VALUE of Liquid Biopsy for Lung Cancer in a Public Healthcare System. J. Thorac. Oncol. 2021, 16, S689. [Google Scholar] [CrossRef]
  34. Leighl, N.B.; Page, R.D.; Raymond, V.M.; Daniel, D.B.; Divers, S.G.; Reckamp, K.L.; Villalona-Calero, M.A.; Dix, D.; Odegaard, J.I.; Lanman, R.B.; et al. Clinical Utility of Comprehensive Cell-Free DNA Analysis to Identify Genomic Biomarkers in Patients with Newly Diagnosed Metastatic Non–Small Cell Lung Cancer. Clin. Cancer Res. 2019, 25, 4691–4700. [Google Scholar] [CrossRef]
Figure 1. Turnaround time of liquid biopsy molecular results, reported in business days.
Figure 1. Turnaround time of liquid biopsy molecular results, reported in business days.
Cancers 16 02505 g001
Figure 2. Distribution of driver mutations in lung cancers (n = 86). * Non-driver mutations include TP53 and PIK3CA, among others.
Figure 2. Distribution of driver mutations in lung cancers (n = 86). * Non-driver mutations include TP53 and PIK3CA, among others.
Cancers 16 02505 g002
Figure 3. Distribution of mutations. (A) Total mutations (n = 22) identified in gastrointestinal cancers (n = 16). (B) Total mutations (n = 18) identified in breast cancers (n = 13).
Figure 3. Distribution of mutations. (A) Total mutations (n = 22) identified in gastrointestinal cancers (n = 16). (B) Total mutations (n = 18) identified in breast cancers (n = 13).
Cancers 16 02505 g003
Figure 4. Utilization patterns of liquid biopsy. Of 124 liquid biopsies, 32 were initiated to inform diagnoses and treatment investigations, 13 to inform resistance and treatment investigations, 11 to inform progression and treatment investigations, 49 to inform treatment investigations only, 6 to inform resistance investigations only, 5 to inform progression investigations only, and 8 for unknown reasons.
Figure 4. Utilization patterns of liquid biopsy. Of 124 liquid biopsies, 32 were initiated to inform diagnoses and treatment investigations, 13 to inform resistance and treatment investigations, 11 to inform progression and treatment investigations, 49 to inform treatment investigations only, 6 to inform resistance investigations only, 5 to inform progression investigations only, and 8 for unknown reasons.
Cancers 16 02505 g004
Table 1. Patient demographic and baseline clinical characteristics.
Table 1. Patient demographic and baseline clinical characteristics.
Characteristicsn = 124 (%)
Age, years
Median (range)67 (23–94)
Sex
Female68 (55)
Male56 (45)
ECOG
018 (15)
166 (53)
224 (19)
38 (6)
42 (2)
Unknown 6 (5)
Stage
III3 (2)
IV114 (92)
Unknown7 (6)
Tumor site
Breast 13 (10)
Gastrointestinal 16 (13)
Other9 (7)
Lung86 (69)
Smoking status (lung cohort)n = 86 (%)
  Non-smoker55 (64)
  Current/former smoker26 (30)
  Unknown5 (6)
Histologic subtype (lung cohort)n = 86 (%)
  Adenocarcinoma77 (90)
  SCC3 (3)
  NOS6 (7)
SCC—squamous cell carcinoma; NOS—not otherwise specified.
Table 2. Comparison of driver alterations between liquid biopsies to matched tissue results.
Table 2. Comparison of driver alterations between liquid biopsies to matched tissue results.
Genomic Alteration Total Cases with Matched TissueMutations Found in Tissue and PlasmaMutations Found in Tissue Only
SNVs, INDELs, CNVs (DNA)nn (%)n (%)
BRAF11-
EGFR31238
ERBB233-
ERBB41-1
KRAS AMPLIFICATION11-
KRAS POINT MUTATION1376
MET AMPLIFICATION1-1
MET SPLICING MUTATION33-
PIK3CA33 -
Total DNA alterations5741 (72%)16 (28%)
FUSIONS (RNA)nn (%)n (%)
ALK422
FGFR111-
MET FUSION312
NTRK1-1
RET1-1
ROS111-
Total RNA alterations115 (46%)6 (55%)
NO ALTERATIONS IDENTIFIED77 (100%)
Total alterations7553 (71%)22 (29%)
SNV—single nucleotide variants; INDELs—insertion/deletion mutations; CNV—copy number variants.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nicholas, C.; Beharry, A.; Bendzsak, A.M.; Bisson, K.R.; Dadson, K.; Dudani, S.; Iafolla, M.; Irshad, K.; Perdrizet, K.; Raskin, W.; et al. Point of Care Liquid Biopsy for Cancer Treatment—Early Experience from a Community Center. Cancers 2024, 16, 2505. https://doi.org/10.3390/cancers16142505

AMA Style

Nicholas C, Beharry A, Bendzsak AM, Bisson KR, Dadson K, Dudani S, Iafolla M, Irshad K, Perdrizet K, Raskin W, et al. Point of Care Liquid Biopsy for Cancer Treatment—Early Experience from a Community Center. Cancers. 2024; 16(14):2505. https://doi.org/10.3390/cancers16142505

Chicago/Turabian Style

Nicholas, Champica, Andrea Beharry, Anna M. Bendzsak, Kassandra R. Bisson, Keith Dadson, Shaan Dudani, Marco Iafolla, Kashif Irshad, Kirstin Perdrizet, William Raskin, and et al. 2024. "Point of Care Liquid Biopsy for Cancer Treatment—Early Experience from a Community Center" Cancers 16, no. 14: 2505. https://doi.org/10.3390/cancers16142505

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop