Next Article in Journal
K63 Ubiquitination of P21 Can Facilitate Pellino-1 in the Context of Chronic Obstructive Pulmonary Disease and Lung Cellular Senescence
Next Article in Special Issue
Tumor-Induced T Cell Polarization by Schwann Cells
Previous Article in Journal
The Tryptophan Catabolite or Kynurenine Pathway in a Major Depressive Episode with Melancholia, Psychotic Features and Suicidal Behaviors: A Systematic Review and Meta-Analysis
Previous Article in Special Issue
Effects of TP53 Mutations and miRs on Immune Responses in the Tumor Microenvironment Important in Pancreatic Cancer Progression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Commentary

Liquid Biopsy and the Translational Bridge from the TIME to the Clinic

Rody School of Medicine at East Carolina University, Greenville, NC 27834, USA
Cells 2022, 11(19), 3114; https://doi.org/10.3390/cells11193114
Submission received: 27 April 2022 / Revised: 9 September 2022 / Accepted: 28 September 2022 / Published: 3 October 2022
(This article belongs to the Special Issue Tumor Immune Microenvironment for Effective Therapy)

Abstract

:
Research and advancing understanding of the tumor immune microenvironment (TIME) is vital to optimize and direct more effective cancer immune therapy. Pre-clinical bench research is vital to better understand the genomic interplay of the TIME and immune therapy responsiveness. However, a vital key to effective translational cancer research is having a bridge of translation to bring that understanding from the bench to the bedside. Without that bridge, research into the TIME will lack an efficient and effective translation into the clinic and cancer treatment decision making. As a clinical oncologist, the purpose of this commentary is to emphasize the importance of researching and improving clinical utility of the bridge, as well as the TIME research itself.

Tumor cell genomics closely intertwine with and direct the TIME compartment’s infiltration and balance of stimulatory and inhibitor immune cells, as well as non-immune stromal components. This sets up an immune ‘hot’ TIME responsive to immune therapy or an immune ‘cold’ TIME not responsive to immune therapy. The TIME is also not a static state. It is a dynamic process changing over time and can be different at different metastatic sites due to progressing metastatic clonal evolution [1].
Tissue biopsies are the standard to assess and understand the TIME. However, assessing the TIME in the clinic can be a far more difficult proposition. Repeat invasive tumor tissue biopsies carry a high procedural cost for tissue acquisition, are fraught with potential complications, cause delays in treatment, and are often not logistically practical to integrate into clinical cancer treatment decision making [2]. Unless evolving knowledge of the TIME can be translated into the clinic, that knowledge will have limited impact identifying effective (and avoiding ineffective) cancer immune treatment as well as missing potential modulation of the TIME to enhance immune therapy benefit.

1. Molecular Tumor Biology Can Reflect the TIME

Cancer is a disease of genomic derangements and instability driving the cancer tumor biology. The underlying molecular tumor biology with targetable driver mutations and fusions as well as actionable tumor co-mutations can reflect differing TIME and immune therapy effectiveness [3,4]. The TIME of EGFR mutation and ALK fusion NSCLC notably lacks CD8 infiltration limiting immune checkpoint blockade benefit [5]. STK11 mutations have strikingly different TIME effects based upon what co-mutations are present. STK11 mutant NSCLC with KRAS co-mutations are associated with increased IL-6, IL-1β, and CXCL7 levels along with neutrophil infiltration, yet decreased T-cell infiltration and function, decreased PD-L1 expression, and decreased stimulator of interferon genes (STING) pathway activation. Studies also point towards associated low intratumoral pH, metabolic restriction, and altered angiogenesis, all leading towards a poor immune therapy benefit [6]. However, the same STK11 mutations with associated TP53 co-mutations but without KRAS mutations, demonstrate increased STING activation and better immune therapy benefit [7]. KRAS mutant NSCLC with TP53 co-mutations also have a completely different TIME with increased IFNγ, PD-L1 expression, and increased T-cell infiltration, supporting an immune therapy benefit [6]. PD-L1 protein, mRNA, and gene amplification are all predictive of ICI responsiveness [8,9,10]. Other mutations such as POLE/POLD1 are associated with a hypermutated state reflecting a favorable immune responsive TIME as do BRCA, SMARCA4, ARID1A, BAP1, and SETD2 mutations [11,12,13,14,15,16]. Whereas STK11, KEAP1, PTEN, β-2 microglobulin mutations, MDM2 amplification, β-catenin pathway alterations, JAK1/2 loss, and oncogenic fusions are often immune therapy resistant [16,17]. Immune therapy hyperprogression in advanced NSCLC has also been associated with STK11 mutations [18]. Even differing EGFR and ERBB2 exon mutations can demonstrate differing immune therapy benefit [19].
Certain other genomic tumor biologies impact the TIME. Microsatellite instability high (MSI-H) has a unique tumor biology and TIME with increased immune cell infiltration, increased neoantigens, increased immune checkpoint expression, increased VEGFR secretion, enhanced STING activation, interferon secretion, and T cell priming, all leading to remarkable immune checkpoint inhibitor (ICI) responsiveness across tissue-site agnostic solid tumors [20,21,22]. A high neoantigen tumor mutational burden (TMB) is also associated with increased ICI responsiveness, albeit to a lesser degree [23].

2. Liquid Biopsy Reflects the Molecular Tumor Biology

Liquid biopsy with plasma next generation sequencing (NGS) is an evolving technology that can identify targetable and actionable molecular tumor biology from circulating tumor DNA (ctDNA) for somatic mutations and RNA (ctRNA) fusions shed from the tumor into the blood [24]. That approach has been highly effective in implementing precision oncology into the clinic and cancer treatment decision making. Although tissue and plasma molecular profiling remain complementary, cancer medicine has entered into a ‘liquid biopsy’ era where simple blood tests are beginning to efficiently identify the underlying tumor molecular biology that can effectively guide treatment.
Tissue molecular testing is fraught with tissue acquisition and spatial heterogeneity limitations. Tissue quantity is insufficient for full molecular testing in nearly half of metastatic non-small lung cancer (NSCLC) cases [25]. Even when available, tissue molecular testing only provides a tumor biology assessment limited to the site sampled and just at that one static point in time. Studies characterizing the clinical utility of liquid biopsy testing, paradoxically show that plasma NGS testing is better than tissue NGS testing for molecular testing. In separate studies with parallel plasma and tissue NGS testing in advanced NSCLC, tissue missed 33–43% of the mutations identified with the complementary testing whereas plasma identified 80–87% of those mutations [26,27]. Plasma NGS testing can also overcome the limiting tissue heterogeneity identifying resistant clones and providing a broader assessment of the evolving tumor biology than a single biopsy site [28]. Most importantly, treatment guided by plasma NGS molecular results have better survival outcomes than treatment based upon tissue molecular testing [29]. This has led the International Association for the Study of Lung Cancer to advocate a ‘plasma first’ molecular testing approach in NSCLC [30]. Liquid biopsies with plasma NGS testing have been highly effective in assessing and reflecting the molecular tumor biology.
However, liquid biopsies are also fraught with limitations. Shedding of ctDNA/RNA from the tumor microenvironment by apoptosis and tumor necrosis is tumor burden, tumor compartment, and tumor genomic microenvironment dependent. The greater the tumor burden and stage, the greater the ctDNA/RNA shedding. In stage I NSCLC 45% will have detectable ctDNA shedding, increasing to 72–75% in lymph node positive stages II/III, and 83% in stage IV [31]. This is typical distribution across a variety of cancers. Non-shedders of ctDNA/RNA have a less aggressive tumor biology with a more favorable prognosis irrespective of stage compared to shedders [32]. In a study of advanced NSCLC patients, multivariate analysis identified visceral metastases, tumor burden, EGFR mutations, and TP53 mutations as independent predictors of increased ctDNA shedding [33]. Higher pathologic stage, nodal metastases, solid adenocarcinoma histology, tumor necrosis, and frequent mitosis were associated with higher ctDNA shedding in resectable NSCLC [34].
Plasma NGS testing can identify ctDNA/RNA alterations, MSI, and TMB associated with ICI sensitive and resistant molecular tumor biologies. Serial ctDNA can also be used to monitor ICI responsiveness with decreasing variant allele fractions and/or clearance predicting durable cancer survival and conversely, progressing disease with increasing ctDNA/RNA [35,36].

3. A Composite Assay Liquid Biopsy Is Needed to Fully Reflect the TIME

However, the TIME is a far more complex entity and is more than mutational tumor biology. ICI responsive immune hot tumors are infiltrated with leukocytes and tumor specific CD8 T-cells. Intratumoral chemokines, IFN-gamma, PD-L1 and, indole 2,3-dioxygenase (IDO) are part of the immune hot TIME responding to ICI therapies. Conversely ICI resistant immune cold tumors express TGF-beta and tumor associated macrophages [37]. Other immune checkpoints such as LAG-3 and TIGIT, and others, may need to be targeted [38,39]. Additionally, the gut microbiome, spatial TIME CD8 T-cell infiltration and tumor cell contact, myeloid inhibition of CD8 T-cells, immunosuppressive myeloid derived suppressor cells, inhibitory T-Reg cells, activated dendritic cells, intratumoral hypoxia, B-cells, cancer associated fibroblasts, all play an important aspect of the TIME [40]. Tumor infiltrating lymphocytes genomic signatures have been shown to be pan-cancer prognostic and predictive of immune therapy benefit [41]. Host inflammatory markers, as simple as a neutrophil-lymphocyte ratio, C-reactive protein levels, and albumin levels, as well as more complex proteomic signatures impact ICI therapy outcomes [42,43]. A composite liquid biopsy of PD-L1with other immune checkpoints, MSI, TMB, ICI sensitive and resistance ctDNA/RNA alterations, immune cellular levels, and incorporating these other TIME, as well as host parameters, is well likely needed to best reflect the TIME and predict ICI treatment benefit.

4. Improving Liquid Biopsy to Better Assess the TIME

The optimal make-up and balance of a composite TIME/ICI predictive liquid biopsy assay is one important research focus. Another aspect of clinically impactful liquid biopsy research is increasing the release of tumor DNA/RNA and other TIME markers to better reflect the TIME. Lack of DNA/RNA shedding, and potential undetectable sub-clones, can limit liquid biopsy assessment of the TIME. However, it is known that certain types of tumor site tissue disruption can increase ctDNA/RNA release. This would further enhance the clinical utility of liquid biopsies in assessing the tumor biology and guiding more effective cancer treatment.
Therapeutic TIME disruption has been shown to enhance ICI effectiveness. Oncolytic virus directly disrupts the TIME [44]. Nanosecond pulsed electric field (PEF) energy delivery has been shown to inhibit tumor growth locally and alter the intratumoral immune cell infiltration and response [45]. Stereotactic radiosurgery (SRS) will disrupt the TIME inducing tumor-specific CD8 T-cells and other TIME changes [46,47].
SRS has been shown to increase release of ctDNA even from early-stage cancers. SRS causes a marked rise in ctDNA at the 24 h mark after the first fraction. In a study of fifteen patients with stage I NSCLC treated by SRS, only 47% had identified ctDNA prior to treatment. However, repeat testing within 24 h of the first SRS fraction showed a median 4.5-fold increase in ctDNA [48]. ctDNA was also increased from baseline upon completion of SRS. Higher levels of ctDNA were noted at 24 h after irradiation than in pre-irradiation samples with a peak at 7 days [49]. There was also a notable increase in targetable ctDNA EGFR mutant allele fractions [50]. One of these therapeutic TIME disruption technologies could certainly be used to enhance ctDNA/RNA and other TIME parameters release into the circulation for liquid biopsy detection.
Whether a lesser physical disruption of the TIME with a simple tissue biopsy achieve this same increase ctDNA/RNA release is not well studied. Nor is there a known optimal time of drawing blood for plasma NGS testing relative to a tissue biopsy to obtain a maximal ctDNA/RNA yield for diagnostic and therapeutic information.
Multiple prostate tissue biopsies have been shown to increase ctDNA release at 60–120 min post-biopsy. In a cohort of thirty-eight patients undergoing and cfDNA testing pre-biopsy and 10, 30, 60, and 120 min post-biopsy, cfDNA peaked at 60–120 min. At 60 min, pre-biopsy median 2.76 ng/mL to 3.62 ng/mL (p = 0.0023) and at 120 min, pre-biopsy 5.1 ng/mL to 7.05 ng/mL (p = 0.0023). Patient-specific somatic mutations were compared pre-biopsy and post-biopsy. The number of reads at the patient-specific mutations increased from 3.9 to 164 times the ratio amounts present circulating before the biopsy. This supports the post-biopsy plasma was enriched with specific ctDNA. The increase was felt to be due to direct physical damage and disruption with cellular breaks and necrosis [51].
Tumor tissue disruption does increase the shedding of ctDNA/RNA from baseline pre-tissue disruption levels. Incorporating these therapeutic tumor disruption modalities could have a dual role of local cancer treatment and enhancing the liquid biopsy identification of the TIME and immune therapy benefit.

5. Improving the Bridge to Translate the TIME into Effective Therapy

Translating the TIME advances from the laboratory bench into the clinic bedside is vital to improving cancer outcomes for patients. The purpose of this commentary was to emphasize the perspective and importance of researching into improving the translational bridge itself and not just focusing on research of the TIME. The quick turn-around times, broadening molecular tumor biology findings, and the extending clinical utility of plasma NGS testing has ushered in the ‘liquid biopsy’ era of cancer medicine. A research focus of this translational bridge is important to improve the translation of the advances and understanding of the TIME into the clinic. Expanded correlation of the genomic tumor biology of the TIME with plasma NGS findings is important. Additional research into expanding the clinical utility development of a composite TIME/ICI responsive liquid biopsy by incorporating other TIME as well as host parameters is needed. Something as simple as research into maximizing the release of ctDNA/RNA and other TIME biomarkers could be important to improve the clinically needed translational bridge. Determining an optimal timing of a liquid biopsy blood draw relative to tissue disruption could also have a significant impact in achieving improved TIME yields.
Research to improve the bridge can be as important as the knowledge transported on it. Without a strong bridge from the bench to the bedside, translating cancer research will not achieve what we all strive for…advancing and optimizing effective cancer immune therapies.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Binnewies, M.; Roberts, E.; Kersten, K.; Chan, V.; Fearon, D.; Merad, M.; Coussins, L.; Gabrilovich, D.; Ostrand-Rosenberg, S.; Hedrick, M.; et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 2018, 24, 541–550. [Google Scholar] [CrossRef] [PubMed]
  2. Kelly, R.; Turner, R.; Chen, Y.-W.; Rigas, J.; Fernandes, A.; Karve, S. Complications and Economic Burden Associated with Obtaining Tissue for Diagnosis and Molecular Analysis in Patients with Non-Small Cell Lung Cancer in the United States. J. Oncol. Pract. 2019, 15, e717–e727. [Google Scholar] [CrossRef] [PubMed]
  3. Shirasawa, M.; Yoshida, T.; Shimoda, Y.; Takayanagi, D.; Shiraishi, K.; Kubo, T.; Mitani, S.; Matsumoto, Y.; Masuda, K.; Shinno, Y.; et al. Differential Immune-related Microenvironment Determines Programmed Cell Death Protein-1/Programmed Death-Ligand 1 Blockade Efficacy in Patients with Advanced NSCLC. J. Thorac. Oncol. 2021, 16, 2078–2090. [Google Scholar] [CrossRef] [PubMed]
  4. Launonen, I.-M.; Lyytikainen, N.; Casado, J.; Anttila, E.; Szabo, A.; Haltia, U.-M.; Jacobson, C.; Lin, J.; Maliga, Z.; Howitt, B.; et al. Single-cell tumor-immune microenvironment of BRCA1/2 mutated high-grade serous ovarian cancer. Nat. Commun. 2022, 13, 835. [Google Scholar] [CrossRef]
  5. Gainor, J.; Shaw, A.; Sequist, L.; Fu, X.; Azzoli, C.; Piotrowska, Z.; Huynh, G.; Zhao, L.; Fulton, L.; Schultz, K.; et al. EGFR Mutations and ALK Rearrangement Are Associated with Low Response Rates to PD-1 Pathway Blockade in Non-Small Cell Lung Cancer: A Retrospective Analysis. Clin. Cancer Res. 2016, 22, 4585–4593. [Google Scholar] [CrossRef] [Green Version]
  6. Skoulidis, F.; Heymach, J. Co-occurring genomic alterations in non-small-cell lung cancer biology and therapy. Nat. Rev. Cancer 2019, 19, 495–509. [Google Scholar] [CrossRef]
  7. Corte, C.; Sen, T.; Gay, C.; Ramkumar, K.; Diao, L.; Cardnell, R.; Rodriguez, B.; Stewart, C.A.; Papadimitrakopoulou, V.; Gibson, L.; et al. STING Pathway Expression Identifies NSCLC with an Immune-Responsive Phenotype. J. Thorac. Oncol. 2020, 15, 777–791. [Google Scholar] [CrossRef]
  8. Mok, T.; Wu, Y.-L.; Kudoba, I.; Kowalski, D.; Cho, B.; Turna, H.; Castro, G., Jr.; Srimuninnimit, V.; Laktionov, K.; Bondarenko, I.; et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1 expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): A randomized, open-label, controlled, phase 3 trial. Lancet 2019, 393, 1819–1830. [Google Scholar] [CrossRef]
  9. Conroy, J.; Pable, S.; Nesline, M.; Glenn, S.; Papanicolau-Sengos, A.; Burgher, B.; Andreas, J.; Giamo, V.; Wang, Y.; Lenzo, F.; et al. Next generation sequencing of PD-L1 for predicting response to immune checkpoint inhibitors. J. Immuno Ther. Cancer 2019, 7, 18. [Google Scholar] [CrossRef] [Green Version]
  10. Goodman, A.; Piccioni, D.; Kato, S.; Boichard, A.; Wang, H.-Y.; Frampton, G.; Lippman, S.; Connelly, C.; Fabrizio, D.; Miller, V.; et al. Prevalence of PDL1 Amplification and Preliminary Response to Immune Checkpoint Blockade in Solid Tumors. JAMA Oncol. 2018, 4, 1237–1244. [Google Scholar] [CrossRef]
  11. Wang, F.; Zhao, Q.; Wang, Y.-N.; Jin, Y.; He, M.-M.; Liu, Z.-X.; Xu, R.-H. Evaluation of POLE and POLD1 Mutations as Biomarkers for Immunotherapy Outcomes Across Multiple Cancer Types. JAMA Oncol. 2019, 5, 1504–1506. [Google Scholar] [CrossRef] [Green Version]
  12. Garmezy, B.; Gheeya, J.; Lin, H.; Huang, Y.; Kim, T.; Jiang, X.; Thein, K.; Pilie, P.; Zeineddine, F.; Wang, W.; et al. Clinical and Molecular Characterization of POLE Mutations as Predictive Biomarkers of Response to Immune Checkpoint Inhibitors in Advanced Cancers. JCO Precis. Oncol. 2022, 6, e2100267. [Google Scholar] [CrossRef]
  13. Zhou, Z.; Li, M. Evaluation of BRCA1 and BRCA2 as Indicators of Response to Immune Checkpoint Inhibitors. JAMA Netw. Open 2021, 4, e217728. [Google Scholar] [CrossRef]
  14. Shen, J.; Ju, Z.; Zhao, W.; Wang, L.; Peng, Y.; Ge, Z.; Nagel, Z.; Zou, J.; Wang, C.; Kapoor, P.; et al. ARID1A deficiency promotes mutability and potentiates therapeutic antitumor immunity unleashed by immune checkpoint blockade. Nat. Med. 2018, 24, 556–562. [Google Scholar] [CrossRef]
  15. Lu, M.; Zhao, B.; Liu, M.; Wu, L.; Li, Y.; Zhai, Y.; Shen, X. Pan-cancer analysis of SETD2 mutation and its association with the efficacy of immunotherapy. Precis. Oncol. 2021, 5, 51. [Google Scholar] [CrossRef]
  16. Fountzilas, E.; Kurzrock, R.; Vo, H.; Tsimberidou, A.-M. Wedding of Molecular Alterations and Immune Checkpoint Blockade: Genomics as a Matchmaker. J. Natl. Cancer Inst. 2021, 113, 1634–1647. [Google Scholar] [CrossRef]
  17. Vidotto, T.; Melo, C.; Castelli, E.; Koti, M.; dos Reis, R.; Squire, J. Emerging role of PTEN loss in evasion of the immune response to tumours. Br. J. Cancer 2020, 122, 1732–1743. [Google Scholar] [CrossRef]
  18. Kim, Y.; Kim, C.; Lee, H.; Lee, S.-H.; Kim, H.; Lee, S.; Cha, H.; Hong, S.; Kim, K.; Seo, S.; et al. Comprehensive Clinical and Genetic Characterization of Hyperprogression Based on Volumetry in Advanced Non-Small Cell Lung Cancer Treated with Immune Checkpoint Inhibitor. J. Thorac. Oncol. 2019, 14, 1608–1618. [Google Scholar] [CrossRef]
  19. Lau, S.; Fares, A.; Le, L.; Mackay, K.; Soberano, S.; Cham, S.; Smith, E.; Ryan, M.; Tsao, M.; Badbury, P.; et al. Subtypes of EGFR- and HER2-Mutation Metastatic NSCLC Influence Response to Immune Checkpoint Inhibitors. Clin. Lung Cancer 2021, 22, 253–259. [Google Scholar] [CrossRef]
  20. Lin, A.; Zhang, J.; Luo, P. Crosstalk Between the MSI Status and Tumor Microenvironment in Colorectal Cancer. Front. Immunol. 2020, 11, 2039. [Google Scholar] [CrossRef]
  21. Narayanan, S.; Kawaguchi, T.; Peng, X.; Qi, Q.; Liu, S.; Yan, L.; Takabe, K. Tumor Infiltrating Lymphocytes and Macrophages Improve Survival in Microsatellite Unstable Colorectal Cancer. Sci. Rep. 2019, 9, 13455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Otto, W.; Macrae, F.; Sierdzinski, J.; Smaga, J.; Krol, M.; Wilinska, E.; Zieniewicz, K. Microsatellite instability and manifestations of angiogenesis in stage IV of sporadic colorectal cancer. Medicine 2019, 98, e13956. [Google Scholar] [CrossRef] [PubMed]
  23. Sha, D.; Jin, Z.; Budczies, J.; Kluck, K.; Stenzinger, A.; Sinicrope, F. Tumor Mutational Burden as a Predictive Biomarker in Solid Tumors. Cancer Discov. 2020, 10, 1808–1825. [Google Scholar] [CrossRef] [PubMed]
  24. Said, R.; Guibert, N.; Oxnard, G.; Tsimberidou, A. Circulating tumor DNA analysis in the era of precision oncology. Oncotarget 2020, 11, 188–211. [Google Scholar] [CrossRef] [Green Version]
  25. Aggarwal, C.; Thompson, J.; Black, T.; Katz, S.; Fan, R.; Yee, S.; Chien, A.; Evans, T.; Baumi, J.; Alley, E.; et al. Clinical Implications of Plasma-Based Genotyping with the Delivery of Personalized Therapy in Metastatic Non-Small Cell Lung Cancer. JAMA Oncol. 2019, 5, 173–180. [Google Scholar] [CrossRef]
  26. Leighl, N.; Page, R.; Raymond, V.; Daniel, D.; Divers, S.; Reckamp, K.; Villalona-Calero, M.; Dix, D.; Lanman, R.; Papadimitrakopoulou, V. 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] [Green Version]
  27. Palmero, R.; Taus, A.; Viteri, S.; Majem, M.; Carcereny, E.; Garde-Noguera, J.; Felipd, E.; Nadal, E.; Malfettone, A.; Sampayo, M.; et al. Biomarker Discovery and Outcomes for Comprehensive Cell-Free Circulating Tumor DNA versus Standard-of-Care Tissue Testing in Advanced Non-Small Cell Lung Cancer. JCO Precis. Oncol. 2021, 5, 93–102. [Google Scholar] [CrossRef]
  28. Parikh, A.; Leshchiner, I.; Elagina, L.; Goyal, L.; Levovitz, C.; Siravegna, G.; Livitz, D.; Rhrissorrakrai, K.; Martin, E.; Van Seventer, E.; et al. Liquid versus tissue biopsy for detecting acquired resistance and tumor heterogeneity in gastrointestinal cancers. Nat. Med. 2019, 25, 1415–1421. [Google Scholar] [CrossRef]
  29. Vidula, N.; Niemierko, A.; Malvarosa, G.; Yuen, M.; Lennerz, J.; Iafrate, A.J.; Wander, S.; Spring, L.; Juric, D.; Isakoff, S.; et al. Tumor Tissue-versus Plasma-based Genotyping for Selection of Matched Therapy and Impact on Clinical Outcomes in Patients with Metastatic Breast Cancer. Clin. Cancer Res. 2021, 27, 3404–3413. [Google Scholar] [CrossRef]
  30. Rolfo, C.; Mack, P.; Scagliotti, G.; Aggarwal, C.; Arcila, M.; Barlesi, F.; Bivona, T.; Diehn, M.; Dive, C.; Dziadziuszko, R.; et al. Liquid Biopsy for Advanced NSCLC: A Consensus Statement from the International Association for the Study of Lung Cancer. J. Thorac. Oncol. 2021, 16, 1647–1662. [Google Scholar] [CrossRef]
  31. Phallen, J.; Sausen, M.; Adleff, V.; Leal, A.; Hruban, C.; White, J.; Anagnostou, V.; Fiksel, J.; Cristiano, S.; Papp, E.; et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci. Transl. Med. 2017, 9, eaan2415. [Google Scholar] [CrossRef] [Green Version]
  32. Vu, P.; Khagi, Y.; Riviere, P.; Goodman, A.; Kurzrock, R. Total Number of Alterations in Liquid Biopsies Is an Independent Predictor for Survival in Patients with Advanced Cancers. JCO Precis. Oncol. 2020, 4, 192–201. [Google Scholar] [CrossRef]
  33. Lam, V.; Zhang, J.; Wu, C.; Tran, H.; Li, L.; Diao, L.; Wang, J.; Rinsurongkawong, W.; Raymond, V.; Lanman, R.; et al. Genotype-Specific Differences in Circulating Tumor DNA Levels in Advanced NSCLC. J. Thorac. Oncol. 2020, 16, 601–609. [Google Scholar] [CrossRef]
  34. Cho, M.-S.; Park, C.; Lee, S.; Park, H. Clinicopathological parameters for circulating tumor DNA shedding in surgically resected non-small cell lung cancer with EGFR or KRAS mutation. PLoS ONE 2020, 15, e0230622. [Google Scholar] [CrossRef] [Green Version]
  35. Vega, D.; Nishimura, K.; Zariffa, N.; Thompson, J.; Hoering, A.; Cilento, V.; Rosenthal, A.; Anagnostou, V.; Baden, J.; Beaver, J.; et al. Changes in Circulating Tumor DNA Reflect Clinical Benefit Across Multiple Studies of Patients with Non-Small-Cell Lung Cancer Treated with Immune Checkpoint Inhibitors. JCO Precis. Oncol. 2022, 6, e2100372. [Google Scholar] [CrossRef]
  36. Gouda, M.; Huang, H.; Piha-Paul, S.; Call, S.G.; Karp, D.; Fu, S.; Naing, A.; Subbiah, V.; Pant, S.; Dustin, D.; et al. Longitudinal Monitoring of Circulating Tumor DNA to Predict Treatment Outcomes in Advanced Cancers. JCO Precis. Oncol. 2022, 6, e2100512. [Google Scholar] [CrossRef]
  37. De Guillebon, E.; Dardenne, A.; Saldmann, A.; Seguier, S.; Tran, T.; Paolini, L.; Lebbe, C.; Tartour, E. Beyond the concept of cold and hot tumors for the development of novel predictive biomarkers and the rational design of immunotherapy combinations. Inter. J. Cancer. 2020, 147, 1509–1518. [Google Scholar] [CrossRef]
  38. Tawbi, H.; Schadendorf, D.; Lipson, E.; Ascierto, P.; Matamala, L.; Gutierrez, E.; Rutkowski, P.; Gogas, H.; Lao, C.; De Menezes, J.; et al. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. N. Engl. J. Med. 2022, 386, 24–34. [Google Scholar] [CrossRef]
  39. Cho, B.; Abreu, D.; Hussein, M.; Cobo, M.; Patel, A.; Secen, N.; Lee, K.; Massuti, B.; Hiret, S.; Yang, J.; et al. Tiragolumab plus atezolizumab versus placebo plus atezolizumab as a first-line treatment for PD-L1 selected non-small-cell lung cancer (CITYSCAPE): Primary and follow-up analyses of a randomized, double-blind, phase 2 study. Lancet Oncol. 2022, 23, 781–792. [Google Scholar] [CrossRef]
  40. Tang, T.; Huang, X.; Zhang, G.; Hong, Z.; Bai, X.; Liang, T. Advantages of targeting the tumor immune microenvironment over blocking immune checkpoint in cancer immunotherapy. Signal Transduct. Target. Ther. 2021, 6, 72. [Google Scholar] [CrossRef]
  41. Ballot, E.; Ladoire, S.; Routy, B.; Truntzer, C.; Ghiringhelli, F. Tumor Infiltrating Lymphocytes Signature as a New Pan-Cancer Predictive Biomarker of Anti PD-1/PD-L1 Efficacy. Cancers 2020, 12, 2418. [Google Scholar] [CrossRef] [PubMed]
  42. Naqash, A.; Stroud, G.; Butt, M.; Dy, G.; Hegde, A.; Muzaffar, M.; Yang, L.; Hafiz, M.; Cherry, C.; Walker, P. Co-relation of overall survival with peripheral blood-based inflammatory biomarkers in advanced stage non-small cell lung cancer treated with anti-programmed cell death-1 therapy: Results from a single institutional database. Acta Oncol. 2017, 57, 867–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Rich, P.; Mitchell, B.; Schaefer, E.; Walker, P.; Dubay, J.; Boyd, J.; Oubre, D.; Page, R.; Khalil, M.; Sinha, S.; et al. Real-world performance of blood-based proteomic profiling in first-line immunotherapy treatment in advanced stage non-small cell lung cancer. J. Immunother. Cancer 2021, 9, e002989. [Google Scholar] [CrossRef] [PubMed]
  44. Hemminki, O.; dos Santos, J.; Hemminki, A. Oncolytic viruses for cancer immunotherapy. J. Hemat. Oncol. 2020, 13, 84. [Google Scholar] [CrossRef]
  45. Zhao, J.; Chen, S.; Zhu, L.; Zhang, L.; Liu, J.; Xu, D.; Tian, G.; Jiang, T. Antitumor Effect and Immune Response of Nanosecond Pulsed Fields in Pancreatic Cancer. Front. Oncol. 2020, 10, 621092. [Google Scholar] [CrossRef] [PubMed]
  46. Gupta, A.; Probst, H.; Vuong, V.; Landshammer, A.; Muth, S.; Yagita, H.; Schwendener, R.; Pruschy, M.; Knuth, A.; van den Broek, M. Radiotherapy Promotes tumor-Specific Effector CD8+ T Cells via Dendritic Cell Activation. J. Immunol. 2012, 189, 558–566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Zhou, P.; Chen, D.; Zhu, B.; Chen, W.; Xie, Q.; Wang, Y.; Tan, Q.; Yuan, B.; Zuo, X.; Huang, C.; et al. Stereotactic Body radiotherapy is Effective in Modifying the Tumor Genome and Tumor Immune Microenvironment in Non-Small Cell Lung Cancer or Lung Metastatic Cancer. Front. Immunol. 2020, 11, 594212. [Google Scholar] [CrossRef]
  48. Chen, E.; Chaudhari, A.; Nabet, B.; Chabon, J.; Merriott, D.; Loo, B.; Alizadeh, A.; Diehn, M. Analysis of Circulating Tumor DNA Kinetics during Stereotactic Ablative Radiation Therapy for Non-Small Cell Lung Cancer. Int. J. Raad Oncol. Biol. Phys. 2018, 102, e767. [Google Scholar] [CrossRef]
  49. Bortolin, M.; Tedeschi, R.; Bidoli, E.; Furlan, C.; Basaglia, G.; Minatel, E.; Gobitti, C.; Franchin, G.; Trovo, M.; De Paoli, P. Cell-free DNA as a prognostic marker in stage I non-small-cell lung cancer patients undergoing stereotactic body radiotherapy. Biomarkers 2015, 20, 422–428. [Google Scholar] [CrossRef]
  50. Kageyama, S.; Nihei, K.; Karasawa, K.; Sawada, T.; Koizumi, E.; Yamaguchi, S.; Kato, S.; Hojo, H.; Motegi, A.; Tsuchiharaa, K.; et al. Radiotherapy increases plasma levels of tumoral cell-free DNA in non-small cell lung cancer patients. Oncotarget 2018, 9, 19368–19378. [Google Scholar] [CrossRef]
  51. Corbetta, M.; Chiereghin, C.; De Simone, I.; Solda, G.; Zuradelli, M.; Giunta, M.; Lughezzani, G.; Buffi, N.; Hurle, R.; Saita, A.; et al. Post-biopsy Cell-Free DNA From Blood: An Open Window on Primary Prostate Cancer Genetics and Biology. Front. Oncol. 2021, 11, 654140. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Walker, P. Liquid Biopsy and the Translational Bridge from the TIME to the Clinic. Cells 2022, 11, 3114. https://doi.org/10.3390/cells11193114

AMA Style

Walker P. Liquid Biopsy and the Translational Bridge from the TIME to the Clinic. Cells. 2022; 11(19):3114. https://doi.org/10.3390/cells11193114

Chicago/Turabian Style

Walker, Paul. 2022. "Liquid Biopsy and the Translational Bridge from the TIME to the Clinic" Cells 11, no. 19: 3114. https://doi.org/10.3390/cells11193114

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