Next Article in Journal
Risankizumab: Daily Practice Experience of High Need Patients
Next Article in Special Issue
Hsp70—A Universal Biomarker for Predicting Therapeutic Failure in Human Female Cancers and a Target for CTC Isolation in Advanced Cancers
Previous Article in Journal
Acute Prosthetic Joint Infections with Poor Outcome Caused by Staphylococcus Aureus Strains Producing the Panton–Valentine Leukocidin
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Clinical Significance of PD-L1 Status in Circulating Tumor Cells for Cancer Management during Immunotherapy

1
Analysis of Circulating Tumor Cells, Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece
2
Department of Internal Medicine, Section of Medical Oncology, National and Kapodistrian University of Athens, Attikon University Hospital, 12462 Athens, Greece
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(6), 1768; https://doi.org/10.3390/biomedicines11061768
Submission received: 31 May 2023 / Revised: 11 June 2023 / Accepted: 16 June 2023 / Published: 20 June 2023
(This article belongs to the Special Issue The Biology of Circulating Tumor Cells 2.0)

Abstract

:
The approval of monoclonal antibodies against programmed death-ligand 1 (PD-L1) and programmed cell death protein (PD1) has changed the landscape of cancer treatment. To date, many immune checkpoint inhibitors (ICIs) have been approved by the FDA for the treatment of metastatic cancer as well as locally recurrent advanced cancer. However, immune-related adverse events (irAEs) of ICIs highlight the need for biomarker analysis with strong predictive value. Liquid biopsy is an important tool for clinical oncologists to monitor cancer patients and administer or change appropriate therapy. CTCs frequently express PD-L1, and this constitutes a clinically useful and non-invasive method to assess PD-L1 status in real-time. This review summarizes all the latest findings about the clinical significance of CTC for the management of cancer patients during the administration of immunotherapy and mainly focuses on the assessment of PD-L1 expression in CTCs.

1. Introduction

Minimally invasive liquid biopsies allow the analysis of tumor elements, such as circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) in body fluids, mainly blood. The liquid biopsy concept launched approximately 10 years ago, has opened new horizons in cancer prevention, diagnosis, early identification of tumor recurrence, molecular characterization of tumors, monitoring of response to treatment, and detection of resistance mechanisms [1]. Clinical applications of CTCs have gained enormous attention over the past few years, despite many limitations in CTC capture by current methodologies [2]. Indeed, CTCs are being evaluated as predictive biomarkers since CTC analysis provides rapid, tumor-specific information that can be repeatedly accessible during follow-up and enables monitoring of response to treatment and early identification of resistance mechanisms.
The Food and Drugs Administration (FDA)-cleared CellSearch platform represents the gold standard for CTC detection and enumeration in the bloodstream [3]. CTC enumeration could provide the primary detection of metastatic cancer in contrast to radiological tests [4]. This creates a huge prospect, especially in the field of early cancer diagnosis. Moreover, one of the main advantages of CTC count using the CellSearch platform is that it enables the stratification of metastatic breast cancer patients (MBC) [5]. Additionally, the implementation of CellSearch allows the detection of CTC clusters that have high metastatic potential and hold great promise for metastatic breast cancer therapy [6].
In recent years, the promising results yielded from the development of CTCs in targeted therapies have paved the way for the implementation of CTCs into the domain of immunotherapy. The most important biomarker for treatment decision-making in the era of immunotherapy is programmed death-ligand 1 (PD-L1) expression, which is assessed by immunohistochemistry in tumor specimens [7]. However, several issues, such as intra-and inter-tumor heterogeneity and expression of PD-L1 in both tumor and immune cells, complicate the accurate measurement of PD-L1 expression in tumor tissues [8]. On the other hand, the possibility of measuring PD-L1 positive CTCs using the CellSearch system was a breakthrough in personalized therapy [9] since CTCs frequently express PD-L1, and this constitutes a clinically useful and non-invasive method to assess PD-L1 status in real-time [9,10].

2. CTC Interaction with Other Cells in the Blood Stream

CTC complex interaction with the immune cells of blood could provide a better understanding of the molecular pathways that are involved, leading to the improvement of therapeutic drugs and reduction of mortality and morbidity associated with cancer [11,12,13]. The activation of platelets represents a critical biological mechanism for metastatic progression since they shield CTCs and protect them from the attack of natural killer (NK) cells or macrophages and facilitate extravasation [14]. Neutrophils assist the metastasis of CTCs and promote tumor development by initiating an angiogenic switch and facilitating the colonization of CTCs [15]. It has also been reported that the abundance of tumor-associated neutrophils (cTAN) in advanced cancer patients contributes to CTC survival by suppressing peripheral leukocyte activation [16]. Moreover, neutrophils represent an important constituent in the formation of CTC clusters [17]. Neutrophil–lung cancer cell interactions are likely to be an important mechanism by which the progression of early malignancy is facilitated [18]. Dendritic cells (DC) cells also play a significant role in the formation of CTC clusters. Recently it was shown that DCs have a strong colocalization effect with CTCs [19]. Moreover, it has been shown that CTCs are associated with abnormalities in peripheral blood DCs in patients with inflammatory breast cancer (IBC). More specifically, IBC patients with ≥5 CTCs have low percentages and impaired function in both subtypes of DCs, indicating that immune cell profiling could add further prognostic value to CTCs in IBC patients [20].
Macrophages prime the premetastatic site and promote tumor cell extravasation, survival, and persistent growth. Macrophages are also immunosuppressive, preventing tumor cell attack by NK and T cells during tumor progression and after recovery from chemo- or immunotherapy [21]. In small cell lung cancer (SCLC), CTCs seem to recruit and “educate” a specific type of macrophages operative in the invasion, immune protection, extravasation, and possibly cachexia [22]. However, macrophages in the liver are major effector cells removing CTCs via antibody-dependent phagocytosis, an immune cell-mediated process preventing liver metastasis [23]. Cancer-associated macrophage-like cells (CAMLs), which are more frequent than CTCs, could provide complementary information for cancer detection and diagnosis [24]. Enumeration of CAMLs using the CellSearch system is related to worse progression-free survival (PFS) and overall survival (OS) compared to patients without CAMLs [25].

3. Clinical Significance of CTCs in Immunotherapy

3.1. Clinical Significance of CTCs and PD-L1+CTCs in Immunotherapy Using CellSearch Platform

CTC enumeration for MBC, metastatic prostate cancer (mPC), and metastatic colorectal cancer (MCC) using CellSearch technology enables the monitoring of cancer patients during therapy. Moreover, this technology captures and identifies tumor cells in the blood that are associated with poor clinical outcomes [26]. CΤC counts have also been associated in several studies with the prognosis of patients undergoing immunotherapy (Table 1). Alama et al. evaluated CTCs in 89 previously treated non-small cell lung cancer (NSCLC) patients receiving nivolumab. In this study, patients with baseline CTC numbers below their median values survived significantly longer [27]. Recent data have highlighted that the metabolic status could affect PD-L1 expression, such as PD-L1 degradation via mitochondria-associated oxidative phosphorylation inhibition [28]. In NSCLC treatment-naïve patients, CTC count variation (ΔCTC) was significantly associated with tumor metabolic response set by the European Organization for Research and Treatment of Cancer (EORTC) criteria. Moreover, elevated CTC count, along with metabolic parameters, were found to be prognostic factors for PFS and OS [29]. Tamminga et al. have shown that CTCs occur in one-third of advanced NSCLC patients, and their presence is of high prognostic and predictive value before and after immunotherapy [30]. In SCLC, that 5-year relative survival rate is extremely low; the use of a much higher cut-off equivalent to 150 CTCs/7.5 mL of whole blood also has clinical utility [31]. In a phase II multicenter adaptive immunotherapy trial of 457 longitudinal liquid biopsies from 104 patients with Metastatic Renal Cell Carcinoma (mRCC), the change over time of CTC enumeration is of prognostic importance [32].
An additional channel in the CellSearch system allows the examination of a fourth molecule of interest beyond the detection of cancer cells of epithelial origin. Establishment of the B7-H1/PD-L1 CTC analysis was performed for the first time by Mazel et al. showing that PD-L1 is frequently expressed on metastatic cells circulating in the blood of hormone receptor-positive, HER2-negative breast cancer patients [9]. This study was followed by several other studies that evaluated the PDL1 status of CTCs in various cancers using CellSearch (Table 1) [33,34,35,36,37,38,39,40].
Nicolazzo et al. showed that in NSCLC patients treated with the programmed cell death protein (PD-1) inhibitor nivolumab at baseline and at 3 months of treatment, the presence of CTCs and the expression of PD-L1 on their surface is associated with poor patients’ outcome. Moreover, 6 months after treatment, patients harboring PD-L1 negative CTCs obtained a clinical benefit, while patients with PD-L1+CTCs all experienced progressive disease, suggesting that the persistence of PD-L1+CTCs might mirror a mechanism of therapy escape [37].
Table 1. Clinical significance of CTCs and PD-L1+-CTCs in immunotherapy using CellSearch platform.
Table 1. Clinical significance of CTCs and PD-L1+-CTCs in immunotherapy using CellSearch platform.
Type of CancerNumber of Samples-PositivityAdditional MarkerTherapyResponseClinical SignificanceRef.
NSCLC89 (91%); baselineNoNivolumabn.aYes;
OS (p = 0.05)
[27]
NSCLC35 (45.7%); baseline
24 (41.7%); 8 weeks
NoNivolumab or PembrolizumabYes;
tumor metabolic response
(p = 0.004)
Yes;
PFS (p < 0.001)
OS (p = 0.024)
[29]
NSCLC30 (36.7%); baselineYes;
PD-L1+CTCs17 (11.8%)
Pembrolizumabn.aYes;
PFS (p = 0.034)
OS (p = 0.023)
[36]
SCLC21 (85.7%); baselineNoChemotherapy or chemotherapy/
immunotherapy
n.aYes;
cut-off ≥ 150 CTCs/7.5 mL
PFS (p = 0.02)
[31]
NSCLC24 (83%); baseline
10 (67%); 3 months
10 (100%); 6 months
Yes;
PD-L1+CTCs
20 (95%); baseline
10 (100%); 3 months
10 (50%); 6 months
NivolumabYes;
PD-L1-CTCs clinical benefit
n.a[37]
NSCLC53 (43.4%)Yes;
PD-L1+CTCs53 (9.4%)
ICIsn.aYes;
CTC countPFS (p = 0.006)
OS (p < 0.001)
PD-L1+CTCs
OS (p = 0.002)
[33]
NSCLC104 (32%); baseline
63 (27%); 4 weeks
NoICIsYes; T0 (p = 0.02) T1 (p < 0.01)Yes;
baseline
PFS (p = 0.05)
OS (p < 0.01)
4 weeks (T1)
PFS (p < 0.01)
OS (p <  0.01)
[30]
NSCLC 39 (15.4%) Yes;
PD-L1+CTCs39 (33.3%)
ICIsn.aYes;
PFS (p = 0.040) OS (p < 0.001)
[35]
mPC10 (50%); pre-ARSI
10 (50%); post-ARSI
10 (40%); mHSPC
Yes;
≥1 PD-L1+CTC10 (60%); pre-ARSI
10 (70%); post-ARSI
10 (40%); mHSPC
Abiraterone, acetate/prednisone or enzalutamiden.an.a[40]
MBC124 (42%)Yes;≥1 PD-L1+CTC
52 (40%)
Chemotherapy, endocrine therapy, targeted therapy n.an.a[34]
MBC16 (100%); ≥1 CTC
16 (81.3%); ≥5 CTC
Yes;
≥1 PD-L1+CTC
16 (68.8%)
n.an.an.a[9]
aUC57 (47.4%); ≥1 CTC 57 (24.6%) ≥5 CTCsYes;
≥1 PD-L1+CTC
16 (62.5%)
Palliative systemic treatmentn.aYes;
≥5 CTC
OS (p = 0.007)
[38]
MCC 51 (41%); ≥1 CTC
51 (33%); >1 CTC
51 (12%); ≥5 CTCs
Yes;
≥1 PD-L1+CTC
4 pts (<1% CTCs weak PD-L1)
n.an.aYes;
≥1 CTC
OS (p = 0.030)
>1 CTC
OS (p < 0.020)
≥5 CTCs
OS (p < 0.0001)
[39]
Sinoquet et al. have shown similar results concerning the worse outcome of PD-L1+CTCs, while PD-L1 expression in tumor tissue failed to prove any prognostic significance [33]. Apart from whole blood, different kinds of biological samples could be analyzed in a CellSearch analyzer, such as pleural fluid specimens. In NSCLC, the non-invasive measurement of PD-L1 expression in pleural EpCAM-positive cells (PECs), using the CellSearch® technology, provides prognostic information and may improve the diagnostic accuracy of malignant pleural effusion (MPE) [41].
In mPC, immunotherapy against immune checkpoint inhibitors (ICIs) seems to be effective. For this purpose, identifying suitable biomarkers could facilitate the selection of the best candidates for this therapy [42]. Expression of PD-L1+ on CTCs in mPC patients during the administration of next-generation AR axis inhibitors is feasible and may enable monitoring of immunotherapy [40]. The expression of PD-L1 on CTCs in blood from patients with advanced urothelial cancer (UC) has also been analyzed. CTC detection and the presence of CTCs with moderate or strong PD-L1 expression are correlated with worse overall survival [38].
The assessment of PD-L1+CTC could also be applied in patients with Merkel cell carcinoma (MCC), which is a rare, aggressive skin cancer with increasing incidence and high mortality rates. Riethdorf et al. show that even though a high prevalence of CTC occurs at first blood collection that is associated with a worse prognosis, the overall frequency of PD-L1 production in CTCs is very low [39].

3.2. Prognostic and Predictive Value of PD-L1+CTCs in Various Types of Cancers

3.2.1. NSCLC

In recent years, immunotherapy has become the first-line treatment for patients with NSCLC, with excellent responses in many patients [43]. However, many patients do not respond to this treatment, so the existence of biomarkers that can direct oncologists to appropriate treatment selection for each patient is essential. According to CheckMate 227, combined immunotherapy has demonstrated durable long-term efficacy benefits over chemotherapy in patients with advanced NSCLC and tumor PD-L1 expression greater than or equal to 1% or less than 1% across nonsquamous and squamous histologies [44]. However, apart from the analysis of PD-L1 in the tissue, its expression can also be studied in CTCs with proven clinical relevance (Table 2 and Table 3) [45]. Ilie et al. reported that PD-L1 expression in CTCs and circulating white blood cells obtained from 106 NSCLC patients correlated with the PD-L1 status in matched tumor-tissue samples [46]. Similar results were also reported by Abdo et al. in a comparative evaluation of PD-L1 in NSCLC patients showing good agreement rates on PD-L1 positivity (TPS ≥ 1%) and high PD-L1 expression (TPS ≥ 50%) [47].
Guibert et al. prospectively analyzed blood samples from 96 advanced-stage NSCLC patients obtained before nivolumab treatment and at the time of disease progression [59]. PD-L1 expression was more frequently observed in CTCs (83%) than in matched tissue samples (41%), and there was no correlation between CTC and tissue PD-L1 expression. Interestingly, a higher pre-treatment PD-L1 positive CTC number was observed in patients that did not respond to nivolumab [PFS < 6 months] [59]. In the same context, a previously mentioned study by Nicolazzo et al. included 24 patients with advanced NSCLC treated with nivolumab and assessed CTCs and CTC PD-L1 expression on blood samples obtained at baseline, 3 months, and 6 months post-treatment [37]. Although at baseline and 3 months post-treatment, detection of CTCs and PD-L1 positivity were associated with a dismal prognosis, at 6 months CTCs were found in all patients included. However, patients with PD-L1 negative CTCs continued to respond to immunotherapy, whereas patients with PD-L1 positive CTCs experienced disease progression, implicating that PD-L1 positivity on CTCs could be a predictive biomarker for early resistance to immunotherapy [37].
The examination of PD-L1 status through sequential biopsies could provide significant prognostic and predictive information due to status changes over time. A longitudinal evaluation of PD-L1 expression of CTCs isolated from NSCLC patients treated with nivolumab was reported by Ikeda et al. CTCs were enriched from 3 mL of peripheral blood using a microcavity array system at baseline and weeks 4, 8, 12, and 24 or until progressive disease. According to this study, PD-L1 expression on CTCs at week 8 has a superior predictive value compared to that at the baseline [55]. In this context, Moran et al. showed that at 18 months, patients showing an increase in PD-L1 expression had better clinical outcomes after ICI, with longer PFS (p = 0.0091) and OS (p = 0.0410) versus patients who did not demonstrate an increase in PD-L1 expression or the no ICI-treated population [48]. A longitudinal analysis was also performed in 47 advanced NSCLC patients receiving pembrolizumab. The results of this study revealed that changes in the PD-L1low subpopulation at an early phase of treatment are importantly related to disease control or resistance to pembrolizumab immunotherapy. Additionally, in patients with partial response, CTC counts were immediately increased at week 3, whereas the PD-L1low CTC rates were decreased [58].
PD-L1 expression presents heterogeneous expression in CTCs and tumor tissues from advanced NSCLC patients. Zhou et al. show that CTCs release a higher detection rate of PD-L1 expression than tumor tissues (53.0% vs. 42.1%). Moreover, NSCLC patients with PD-L1− on tissues but PD-L1+ on CTCs could still benefit from ICI therapy, while co-identification of PD-L1+CTCs or PD-L1+ tissues may help to identify patients who would benefit from immunotherapy [56].
Enough data support the fact that upon disease progression, NSCLC patients demonstrate an increase in PD-L1+CTCs, while no change or a decrease in PD-L1+CTCs is observed in responding patients [57]. Additionally, the increase of PD-L1+CTCs might indicate resistance toward PD-1/PD-L1 inhibitors. Similar results were shown by Sinoquet et al., where OS was significantly worse in NSCLC patients with PD-L1-CTCs and particularly in patients with PD-L1+CTCs compared with patients without CTCs. Moreover, the presence of PD-L1+CTC correlated with the absence of gene alterations in tumor tissue and with poor prognosis-related biological variables (anemia, hyponatremia, and increased lactate dehydrogenase) [33].
PD-L1 expression has also been studied in groups of patients receiving other types of treatment besides immunotherapy. Wang et al. studied gene expression of PD-L1 in CTCs isolated before, during, and after radiation or chemoradiation using a microfluidic chip. PD-L1 mRNA was highly expressed in patients who had disease progression within 9 months compared to those who had stable disease for 9 months or more, indicating that radiation therapy induces PD-L1 expression in CTCs [50].
The dynamic probability of PD-L1 as a surrogate marker has also been analyzed in multiple basket studies. Tan et al., in a study involving one hundred fifty-five patients with different advanced cancers, showed that the reduction in CTC counts and ratios of PD-L1-positive CTCs and PD-L1-high CTCs reflect a beneficial response to PD-1/PD-L1 inhibitors. In this study, patients with PD-L1-high CTCs had significantly longer PFS (4.9 vs. 2.2 months, p < 0.0001) and OS (16.1 vs. 9.0 months, p = 0.0235) than those without PD-L1-high CTCs [49]. Recently, a meta-analysis was reported, including results from 30 eligible studies (32 cohorts, 1419 cancer patients) about the prognostic significance of PD-L1 expression on CTCs in various cancers. The overall results from this meta-analysis showed that pre-treatment PD-L1+CTCs might predict better survival for patients receiving ICI treatment but worse survival for patients receiving other therapies. In addition, post-treatment PD-L1+CTCs were correlated with worse survival in cancers [60].

3.2.2. HNSCC

In head and neck squamous cell carcinoma (HNSCC), the administration of immunotherapies has led to a response rate equivalent to 15–20%. However, ICIs have been approved for recurrent and metastatic (R/M) HNSCC patients as a first- and second-line therapy [61,62]. Recent data have revealed that CTC analysis is very promising in HNSCC [63,64,65]. However, studies on the clinical utility of PD-L1-positive CTCs are limited. In a prospective study including 23 HNC patients (Stages I–IV), PD-L1 status in CTCs was examined and correlated to patients’ survival. CTC enrichment was performed using the ClearCell FX system, which separates cells based on size (>14 µm) and deformability parameters. CTC immunophenotyping revealed that more than half of the patients (54.4%) appear to express PD-L1. Moreover, patients with CTC-positive counts had shorter PFS than patients with the absence of CTCs (hazard ratio [HR]: 4.946; 95% confidence interval [CI]: 1.571–15.57; p = 0.0063), and the PD-L1 positivity in the CTCs was found to be significant ([HR]: 5.159; 95% [CI]: 1.011–26.33; p = 0.0485) [51].
A highly sensitive, specific, and robust RT-qPCR assay for PD-L1 mRNA expression in EpCAM(+) CTCs has been developed by Strati et al. for the detection of PD-L1 overexpression in CTC. This prospective study enrolled 113 locally advanced HNSCC patients treated with curative intent at baseline, after two cycles of induction chemotherapy (week 6), and at the end of concurrent chemoradiotherapy (week 15). The findings of this study showed that patients with CTCs overexpressing PD-L1 at the end of treatment had worse outcomes (PFS; p = 0.001, OS; p < 0.001), while its absence was strongly associated with complete response (95% CI = 2.76–92.72, p = 0.002) [10].

3.2.3. Prostate Cancer

Immunotherapy represents a promising therapeutic option for the cure of prostate cancer patients [66]. In a phase II study, Boudadi et al. enrolled 16 patients with metastatic prostate cancer and AR-V7 expressing CTCs, that were prospectively treated with a combination of nivolumab and ipilimumab. Using targeted next-generation sequencing (NGS) in both pre-treatment tumor samples and CTCs, the authors found that high CTC phenotypic heterogeneity using the Shannon index was associated with improved response to combination immunotherapy. In addition, patients with defects in DNA repair genes (assessed by NGS in tumor biopsies or cell-free DNA in the case of no tissue availability) had higher CTC heterogeneity [67]. Zhang et al. performed CTC analysis for immune checkpoint ligands expression in men with mPC. Three cohorts of patients were enrolled, receiving different combinations of new-generation hormone therapy. High heterogeneity of immune checkpoint expression on CTCs was revealed across different disease states [40].

3.2.4. Breast Cancer

Mazel and co-authors were the first to report the expression of PD-L1 on CTCs of patients with ER(+) HER2(−) breast cancer (BC) [9]. Interestingly, this study showed remarkable heterogeneity regarding PD-L1 expression in CTCs among the PD-L1 positive patients (11 out of 16, 68.8%). Schott et al. detected PD-L1 and PD-L2 expression on CTCs derived from blood samples of 128 patients with breast, prostate, lung, and colorectal cancer. In this study, patients with MBC had significantly more PD-L1 positive CTCs compared to patients with non-metastatic disease [68]. In addition, in one patient with MBC treated with combination immunotherapy (nivolumab/ipilimumab), the proportion of PD-L1 positive CTCs declined after the first and second dose of immunotherapy, whereas it increased following drug interruption, despite the persistently low level of CTCs.
Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer that molecular targeted therapies are lacking. Immunotherapy has been included as standard care for stage II-III TNBC [69]. Vardas et al. studied a panel of ICIs, including PD-L1, in sixty-four BC patients with TNBC and thirty-one with luminal A or B of early and metastatic disease. Among BC subtypes, the phenotype of PD-L1+CD45CK+ was higher in TNBC compared to luminal patients. Furthermore, among TNBC patients, there was an association of the phenotype PD-L1+CD45CK+ with a shorter OS (7.6 vs. 53.8 months; log-rank p < 0.001, HR = 8.7) [54]

3.2.5. Melanoma

The high immunogenicity of melanoma cancer makes immunotherapy one of the most effective treatment strategies [70]. Molecular characterization of circulating melanoma cells provides monitoring of the early response to immunotherapy [71]. Khattak MA et al. performed a longitudinal analysis of PD-L1 expression on CTCs in patients with metastatic melanoma receiving pembrolizumab prior to treatment and 6–12 weeks after initiation of therapy. PD-L1 positivity was prevalent in a high percentage of CTCs (64%) derived from melanoma patients. Moreover, patients with one or more PD-L1+CTCs had a higher response rate to pembrolizumab, as well as longer PFS compared with patients with PD-L1-CTCs (26.6 months vs. 5.5 months; p = 0.018) [52].

3.2.6. Other Types of Cancers (Genitourinary Cancer, Bladder Cancer, Hepatocellular Cancer)

Chalfin et al. evaluated the T-cell counts and CTC morphologic features of metastatic genitourinary cancer patients receiving combination immunotherapy at baseline and on therapy at cycle 2 and cycle 3. Five distinct morphologic subtypes were identified by calculating the Shannon Index, and increasing CTC heterogeneity during therapy administration was associated with worse OS. Moreover, patients with CTCs > 4, specific CTC morphologic subtypes, PD-L1+, and low CD4 and CD8 T-cell counts had shorter survival [72].
Immunological response to bladder cancer is well conserved, and PD-L1 expression is differentiated between high-grade and low-grade cancers [73]. Morelli et al. show that 90% of non-muscle-invasive bladder cancer (NMIBC) patients have detectable CTCs, with a median CTC count of about four. A significant correlation between high PD-L1 and reduced recurrence-free survival (RFS) makes NMIBC patients’ ideal candidates for systemic approaches with ICIs [74].
In hepatocellular cancer (HCC), Su et al. investigated the predictive value of PD-L1 expression on CTCs in patients receiving PD-1 inhibitors combined with radiotherapy and antiangiogenic therapy. The count of PD-L1+CTCs was found to be an independent predictive biomarker of OS, and the objective response was more likely to be achieved in patients with a dynamic decrease in PD-L1+CTC counts at 1 month after treatment [53].

4. Immunotherapeutics on CTCs

CTCs acquire key properties required for metastatic spread and constitute an intermediate stage of metastasis [75]. They exist in the bloodstream as single cells or clusters of cells that are oligoclonal precursors of breast cancer metastasis [76]. The discovery of their molecular traits could facilitate the identification of targeted therapies [77]. Viable CTCs could be subjected to a dormant state through the immune-escape mechanism of CD47 upregulation [78,79]. Simultaneously blocking CD274 (programmed death ligand 1, PD-L1, or B7-H1) and CD47 checkpoints on CTCs by corresponding antibodies enhances the inhibition of tumor growth [80].
NK cells are of major importance in host immunity against cancer. Several different approaches to NK-based immunotherapy have been reported [81]. Allogeneic NK cells immunotherapy for recurrent breast cancer [82] and NSCLC [83] decrease CTC levels, which reflects the efficacy of treatment. A decrease in the number of CTCs is also an indication of oncolytic viral immunotherapy (Olvi-Vec). In an open-label phase 1b trial intraperitoneal, Olvi-Vec was given as monotherapy in two consecutive daily doses in 12 patients with platinum-resistant or refractory ovarian cancer. Immune activation was demonstrated from virus-enhanced tumor infiltration of CD8+ T-cells and activation of tumor-specific T-cells in peripheral blood, while at the same time, CTCs were diminished in 6/8 (75%) of baseline-positive patients [84]. A single-center prospective study demonstrated the short-term safety and efficacy of irreversible electroporation (IRE) combined with allogenic NK cell immunotherapy for unresectable primary liver cancer (PLCs). The combination therapy of IRE and NK cell immunotherapy significantly reduced CTCs and increased immune function and Karnofsky’s performance status. Moreover, PFS and OS were significantly improved in the IRE–NK group, demonstrating the synergistic effect of these two therapies [85]. Recent studies have also shown that exosomes derived from NK cells also exhibit antitumor properties. Kang YT et al. developed a streamlined microfluidic approach to on-chip biogenesis and harvest of natural killer cell-derived exosomes through comprehensive studies using NK cell lines and clinical samples from lung cancer patients. Circulating NK cell-derived exosomes have a cytotoxic effect against in-house patient-derived expanded CTC lines [86].
TAMs are the most frequent immune cells within the tumor microenvironment [87]. Sialic acid-modified EPI-loaded liposomes (EPI-SL) inhibit breast cancer metastasis by targeting TAMs and CTCs. A basic constituent of EPI-SL is the ligand of SA-CH, composed of sialic acid (SA) and cholesterol (CH). This is critical since SA-CH can directly bind to selectin, which is highly expressed on the surface of CTCs and effectively target and captures CTCs [88]. A HER2/neu vaccine-based immunotherapy for breast cancer has been reported in a pilot study by Stojadinovic A. et al. HER2/neu represents an attractive molecular target as an anticancer vaccine in breast cancer since it is overexpressed in up to 30% of breast cancers. E75+GMCSF vaccination was applied in 16 patients with HER2/neu-expressing primary breast cancer, while thirteen of the 16 patients (81.3%) had at least one HER2/neu+CTC (mean: 2.1 ± 0.1 CTC/20 mL) in the peripheral blood. After vaccination, a reduction in CTC/20 mL (pre-vaccination 3.9 ± 1.5 vs. postvaccination 0.7 ± 0.4, p = 0.077) and HER2/neu+CTC/20 mL (pre-vaccination 2.8 ± 1.0 vs. postvaccination 0.5 ± 0.2, p = 0.048) was demonstrated [89].
In-vitro experiments have shown that immune activation of the monocyte-derived dendritic cells (Mo-DCs) using patients’ own CTCs is feasible. Kolostova K. et al. performed a co-culture of mature Mo-DCs (mMo-DCs) and autologous non-target blood cells (NTBCs). The activation effect of mature Mo-DCs on T-cell activation was monitored using multimarker gene expression profiling. Moreover, mMo-DCs might play a significant role in the PD-L1/PD1 regulatory axis since an elevated gene expression of PD-L1 was observed [90].

5. Future Perspectives and Conclusions

Liquid biopsy represents a novel, non-invasive approach for detecting and monitoring cancer through the analysis of its biological components, such as CTCs. The main challenge of the liquid biopsy era is the primary detection of minimal residual disease (MRD), where cancer cells, disseminated from the primary tumor, are non-detectable with conventional clinical or radiological tests, increasing the probability of new tumors formation of high metastatic potential. Sensitive and specific isolation and detection of CTCs is very important, especially in the case where surgical removal of the tumor is difficult. In this case, the information from the tumor cannot be available, and thus, oncologists do not have the proper guidance for the correct administration of targeted therapy to the patients.
Immunotherapy activates the body’s immune system to destroy cancer cells by enhancing the recognition ability of immune cells to the surface antigens of tumor cells, achieving their elimination. PD-L1 is a critical immune checkpoint protein that binds to PD-1 in T cells. ICIs are blocking the PD-1/PDL-1 interaction enabling immune system attack and sequentially destroying the cancer cells. That being said, it highlights the necessity of technologies that can accurately determine and assess the status of PD-L1 biomarkers and guide clinical oncologists as to whether cancer patients are suitable for immunotherapy. However, larger clinical studies are needed to be performed for the evaluation of the PDL1 status of CTCs and the integration of the PD-L1-CTC test into daily clinical practice.

Author Contributions

Conceptualization, A.S.; investigation, A.S. and P.E.; writing—original draft preparation, AS., P.E., E.L. and A.P.; writing—review and editing, A.S.; supervision, A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This study has been financially supported by the European Union and Greek national funds through the Operational Program: Competitiveness, Entrepreneurship and Innovation, under the call RESEARCH—CREATE—INNOVATE (project code: T1RCI-02935).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Full NameAbbreviationFull NameAbbreviation
Programmed death-ligand 1PD-L1Programmed cell death proteinPD1
Circulating Tumor CellsCTCsFood and Drugs AdministrationFDA
Circulating Tumor DNActDNAMetastatic breast cancerMBC
Natural KillerNKImmune checkpoint inhibitorsICIs
Dendritic cellsDCTumor-associated neutrophilscTAN
Small Cell Lung CancerSCLCInflammatory breast cancerIBC
Progression Free SurvivalPFSCancer-associated macrophage-like cellsCAMLs
CTC count variationΔCTCOverall survivalOS
Pleural EpCAM-positive cellsPECsMetastatic prostate cancermPC
Malignant pleural effusionMPEMetastatic colorectal cancerMCC
Urothelial CancerUCNon-small cell lung cancerNSCLC
Merkel Cell CarcinomaMCCEuropean Organization for Research and Treatment of CancerEORTC
Recurrence-Free SurvivalRFSMetastatic renal cell carcinomamRCC
Hepatocellular cancerHCCHead and neck squamous cell carcinomaHNSCC
Irreversible electroporationIRENext-generation sequencingNGS
Sialic acidSATriple negative breast cancerTNBC
CholesterolCHNon-muscle-invasive bladder cancerNMIBC
Mature Mo-DCsmMo-DCsSialic Acid-Modified EPI-Loaded LiposomesEPI-SL
Non-target blood cellsNTBCsMonocyte-derived dendritic cellsMo-DCs
Oncolytic viral immunotherapyOlvi-VecMinimal residual diseaseMRD
Immune-related adverse eventsirAEs

References

  1. Alix-Panabières, C.; Pantel, K. Liquid Biopsy: From Discovery to Clinical Application. Cancer Discov. 2021, 11, 858–873. [Google Scholar] [CrossRef] [PubMed]
  2. Economopoulou, P.; Kotsantis, I.; Kyrodimos, E.; Lianidou, E.S.; Psyrri, A. Liquid Biopsy: An Emerging Prognostic and Predictive Tool in Head and Neck Squamous Cell Carcinoma (HNSCC). Focus on Circulating Tumor Cells (CTCs). Oral. Oncol. 2017, 74, 83–89. [Google Scholar] [CrossRef]
  3. Cristofanilli, M.; Budd, G.T.; Ellis, M.J.; Stopeck, A.; Matera, J.; Miller, M.C.; Reuben, J.M.; Doyle, G.V.; Allard, W.J.; Terstappen, L.W.M.M.; et al. Circulating Tumor Cells, Disease Progression, and Survival in Metastatic Breast Cancer. N. Engl. J. Med. 2004, 351, 781–791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Giordano, A.; Cristofanilli, M. CTCs in Metastatic Breast Cancer. Recent Results Cancer Res. 2012, 195, 193–201. [Google Scholar] [CrossRef] [PubMed]
  5. Cristofanilli, M.; Pierga, J.Y.; Reuben, J.; Rademaker, A.; Davis, A.A.; Peeters, D.J.; Fehm, T.; Nolé, F.; Gisbert-Criado, R.; Mavroudis, D.; et al. The Clinical Use of Circulating Tumor Cells (CTCs) Enumeration for Staging of Metastatic Breast Cancer (MBC): International Expert Consensus Paper. Crit. Rev. Oncol. Hematol. 2019, 134, 39–45. [Google Scholar] [CrossRef] [PubMed]
  6. Piñeiro, R.; Martínez-Pena, I.; López-López, R. Relevance of CTC Clusters in Breast Cancer Metastasis. Adv. Exp. Med. Biol. 2020, 1220, 93–115. [Google Scholar] [CrossRef]
  7. Zhou, Z.; Liu, Y.; Jiang, X.; Zheng, C.; Luo, W.; Xiang, X.; Qi, X.; Shen, J. Metformin Modified Chitosan as a Multi-Functional Adjuvant to Enhance Cisplatin-Based Tumor Chemotherapy Efficacy. Int. J. Biol. Macromol. 2023, 224, 797–809. [Google Scholar] [CrossRef]
  8. Ilie, M.; Long-Mira, E.; Bence, C.; Butori, C.; Lassalle, S.; Bouhlel, L.; Fazzalari, L.; Zahaf, K.; Lalvée, S.; Washetine, K.; et al. Comparative Study of the PD-L1 Status between Surgically Resected Specimens and Matched Biopsies of NSCLC Patients Reveal Major Discordances: A Potential Issue for Anti-PD-L1 Therapeutic Strategies. Ann. Oncol. 2016, 27, 147–153. [Google Scholar] [CrossRef]
  9. Mazel, M.; Jacot, W.; Pantel, K.; Bartkowiak, K.; Topart, D.; Cayrefourcq, L.; Rossille, D.; Maudelonde, T.; Fest, T.; Alix-Panabi Eres, C. Frequent Expression of PD-L1 on Circulating Breast Cancer Cells. Mol. Oncol. 2015, 9, 1773–1782. [Google Scholar] [CrossRef] [Green Version]
  10. Strati, A.; Koutsodontis, G.; Papaxoinis, G.; Angelidis, I.; Zavridou, M.; Economopoulou, P.; Kotsantis, I.; Avgeris, M.; Mazel, M.; Perisanidis, C.; et al. Prognostic Significance of PD-L1 Expression on Circulating Tumor Cells in Patients with Head and Neck Squamous Cell Carcinoma. Ann. Oncol. 2017, 28, 1923–1933. [Google Scholar] [CrossRef]
  11. Bauer, A.T.; Gorzelanny, C.; Gebhardt, C.; Pantel, K.; Schneider, S.W. Interplay between Coagulation and Inflammation in Cancer: Limitations and Therapeutic Opportunities. Cancer Treat. Rev. 2022, 102, 102322. [Google Scholar] [CrossRef] [PubMed]
  12. Dotse, E.; Lim, K.H.; Wang, M.; Wijanarko, K.J.; Chow, K.T. An Immunological Perspective of Circulating Tumor Cells as Diagnostic Biomarkers and Therapeutic Targets. Life 2022, 12, 323. [Google Scholar] [CrossRef] [PubMed]
  13. Garrido-Navas, C.; de Miguel-Pérez, D.; Exposito-Hernandez, J.; Bayarri, C.; Amezcua, V.; Ortigosa, A.; Valdivia, J.; Guerrero, R.; Puche, J.L.G.; Lorente, J.A.; et al. Cooperative and Escaping Mechanisms between Circulating Tumor Cells and Blood Constituents. Cells 2019, 8, 1382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Cheng, X.; Zhang, H.; Hamad, A.; Huang, H.; Tsung, A. Surgery-Mediated Tumor-Promoting Effects on the Immune Microenvironment. Semin. Cancer Biol. 2022, 86, 408–419. [Google Scholar] [CrossRef]
  15. Tao, L.; Zhang, L.; Peng, Y.; Tao, M.; Li, L.; Xiu, D.; Yuan, C.; Ma, Z.; Jiang, B. Neutrophils Assist the Metastasis of Circulating Tumor Cells in Pancreatic Ductal Adenocarcinoma: A New Hypothesis and a New Predictor for Distant Metastasis. Medicine 2016, 95, e4932. [Google Scholar] [CrossRef]
  16. Zhang, J.; Qiao, X.; Shi, H.; Han, X.; Liu, W.; Tian, X.; Zeng, X. Circulating Tumor-Associated Neutrophils (CTAN) Contribute to Circulating Tumor Cell Survival by Suppressing Peripheral Leukocyte Activation. Tumor Biol. 2016, 37, 5397–5404. [Google Scholar] [CrossRef]
  17. Szczerba, B.M.; Castro-Giner, F.; Vetter, M.; Krol, I.; Gkountela, S.; Landin, J.; Scheidmann, M.C.; Donato, C.; Scherrer, R.; Singer, J.; et al. Neutrophils Escort Circulating Tumour Cells to Enable Cell Cycle Progression. Nature 2019, 566, 553–557. [Google Scholar] [CrossRef]
  18. Spicer, J.D.; McDonald, B.; Cools-Lartigue, J.J.; Chow, S.C.; Giannias, B.; Kubes, P.; Ferri, L.E. Neutrophils Promote Liver Metastasis via Mac-1-Mediated Interactions with Circulating Tumor Cells. Cancer Res. 2012, 72, 3919–3927. [Google Scholar] [CrossRef] [Green Version]
  19. Zeng, X.; Wei, D.; Wei, X. Background Modeling Method to Identify Interactions Between Circulating Tumor Cells and Dendritic Cells. In Proceedings of the 2018 40th Annual International Conference of the IEEE Engineering in Medicine and Biology Society (EMBC), Honolulu, HI, USA, 18–21 July 2018; Volume 2018, pp. 806–809. [Google Scholar] [CrossRef]
  20. Mego, M.; Gao, H.; Cohen, E.N.; Anfossi, S.; Giordano, A.; Tin, S.; Fouad, T.M.; Giorgi, U.D.; Giuliano, M.; Woodward, W.A.; et al. Circulating Tumor Cells (CTCs) Are Associated with Abnormalities in Peripheral Blood Dendritic Cells in Patients with Inflammatory Breast Cancer. Oncotarget 2017, 8, 35656–35668. [Google Scholar] [CrossRef] [Green Version]
  21. Noy, R.; Pollard, J.W. Tumor-Associated Macrophages: From Mechanisms to Therapy. Immunity 2014, 41, 49–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Hamilton, G.; Rath, B. Circulating Tumor Cell Interactions with Macrophages: Implications for Biology and Treatment. Transl. Lung Cancer Res. 2017, 6, 418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Gül, N.; Babes, L.; Kubes, P.; Van Egmond, M. Macrophages in the Liver Prevent Metastasis by Efficiently Eliminating Circulating Tumor Cells after Monoclonal Antibody Immunotherapy. Oncoimmunology 2014, 3, e28441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Tang, C.M.; Adams, D.L. Clinical Applications of Cancer-Associated Cells Present in the Blood of Cancer Patients. Biomedicines 2022, 10, 587. [Google Scholar] [CrossRef] [PubMed]
  25. Mu, Z.; Wang, C.; Ye, Z.; Rossi, G.; Sun, C.; Li, L.; Zhu, Z.; Yang, H.; Cristofanilli, M. Prognostic Values of Cancer Associated Macrophage-like Cells (CAML) Enumeration in Metastatic Breast Cancer. Breast Cancer Res. Treat. 2017, 165, 733–741. [Google Scholar] [CrossRef]
  26. Swennenhuis, J.F.; van Dalum, G.; Zeune, L.L.; Terstappen, L.W.M.M. Improving the CellSearch® System. Expert. Rev. Mol. Diagn. 2016, 16, 1291–1305. [Google Scholar] [CrossRef] [Green Version]
  27. Alama, A.; Coco, S.; Genova, C.; Rossi, G.; Fontana, V.; Tagliamento, M.; Dal Bello, M.G.; Rosa, A.; Boccardo, S.; Rijavec, E.; et al. Prognostic Relevance of Circulating Tumor Cells and Circulating Cell-Free DNA Association in Metastatic Non-Small Cell Lung Cancer Treated with Nivolumab. J. Clin. Med. 2019, 8, 1011. [Google Scholar] [CrossRef] [Green Version]
  28. Zhou, Z.; Liu, Y.; Song, W.; Jiang, X.; Deng, Z.; Xiong, W.; Shen, J. Metabolic Reprogramming Mediated PD-L1 Depression and Hypoxia Reversion to Reactivate Tumor Therapy. J. Control. Release 2022, 352, 793–812. [Google Scholar] [CrossRef]
  29. Castello, A.; Carbone, F.G.; Rossi, S.; Monterisi, S.; Federico, D.; Toschi, L.; Lopci, E. Circulating Tumor Cells and Metabolic Parameters in NSCLC Patients Treated with Checkpoint Inhibitors. Cancers 2020, 12, 487. [Google Scholar] [CrossRef] [Green Version]
  30. Tamminga, M.; De Wit, S.; Hiltermann, T.J.N.; Timens, W.; Schuuring, E.; Terstappen, L.W.M.M.; Groen, H.J.M. Circulating Tumor Cells in Advanced Non-Small Cell Lung Cancer Patients Are Associated with Worse Tumor Response to Checkpoint Inhibitors. J. Immunother. Cancer 2019, 7, 173. [Google Scholar] [CrossRef] [Green Version]
  31. Mondelo-Macía, P.; García-González, J.; Abalo, A.; Mosquera-Presedo, M.; Aguín, S.; Mateos, M.; López-López, R.; León-Mateos, L.; Muinelo-Romay, L.; Díaz-Peña, R. Plasma Cell-Free DNA and Circulating Tumor Cells as Prognostic Biomarkers in Small Cell Lung Cancer Patients. Transl. Lung Cancer Res. 2022, 11, 1995–2009. [Google Scholar] [CrossRef] [PubMed]
  32. Bootsma, M.; Mckay, R.R.; Emamekhoo, H.; Bade, R.M.; Schehr, J.L.; Mannino, M.C.; Singh, A.; Wolfe, S.K.; Schultz, Z.D.; Sperger, J.; et al. Longitudinal Molecular Profiling of Circulating Tumor Cells in Metastatic Renal Cell Carcinoma. J. Clin. Oncol. 2022, 40, 3633–3641. [Google Scholar] [CrossRef]
  33. Sinoquet, L.; Jacot, W.; Gauthier, L.; Pouderoux, S.; Viala, M.; Cayrefourcq, L.; Quantin, X.; Alix-Panabières, C. Programmed Cell Death Ligand 1-Expressing Circulating Tumor Cells: A New Prognostic Biomarker in Non-Small Cell Lung Cancer. Clin. Chem. 2021, 67, 1503–1512. [Google Scholar] [CrossRef]
  34. Darga, E.P.; Dolce, E.M.; Fang, F.; Kidwell, K.M.; Gersch, C.L.; Kregel, S.; Thomas, D.G.; Gill, A.; Brown, M.E.; Gross, S.; et al. PD-L1 Expression on Circulating Tumor Cells and Platelets in Patients with Metastatic Breast Cancer. PLoS ONE 2021, 16, e0260124. [Google Scholar] [CrossRef]
  35. Dall’Olio, F.G.; Gelsomino, F.; Conci, N.; Marcolin, L.; De Giglio, A.; Grilli, G.; Sperandi, F.; Fontana, F.; Terracciano, M.; Fragomeno, B.; et al. PD-L1 Expression in Circulating Tumor Cells as a Promising Prognostic Biomarker in Advanced Non-Small-Cell Lung Cancer Treated with Immune Checkpoint Inhibitors. Clin. Lung Cancer 2021, 22, 423–431. [Google Scholar] [CrossRef]
  36. Mondelo-Macía, P.; García-González, J.; León-Mateos, L.; Anido, U.; Aguín, S.; Abdulkader, I.; Sánchez-Ares, M.; Abalo, A.; Rodríguez-Casanova, A.; Díaz-Lagares, Á.; et al. Clinical Potential of Circulating Free DNA and Circulating Tumour Cells in Patients with Metastatic Non-Small-Cell Lung Cancer Treated with Pembrolizumab. Mol. Oncol. 2021, 15, 2923–2940. [Google Scholar] [CrossRef]
  37. Nicolazzo, C.; Raimondi, C.; Mancini, M.; Caponnetto, S.; Gradilone, A.; Gandini, O.; Mastromartino, M.; Del Bene, G.; Prete, A.; Longo, F.; et al. Monitoring PD-L1 Positive Circulating Tumor Cells in Non-Small Cell Lung Cancer Patients Treated with the PD-1 Inhibitor Nivolumab. Sci. Rep. 2016, 6, 31726. [Google Scholar] [CrossRef] [PubMed]
  38. Bergmann, S.; Coym, A.; Ott, L.; Soave, A.; Rink, M.; Janning, M.; Stoupiec, M.; Coith, C.; Peine, S.; von Amsberg, G.; et al. Evaluation of PD-L1 Expression on Circulating Tumor Cells (CTCs) in Patients with Advanced Urothelial Carcinoma (UC). Oncoimmunology 2020, 9, 1738798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Riethdorf, S.; Hildebrandt, L.; Heinzerling, L.; Heitzer, E.; Fischer, N.; Bergmann, S.; Mauermann, O.; Waldispühl-Geigl, J.; Coith, C.; Schön, G.; et al. Detection and Characterization of Circulating Tumor Cells in Patients with Merkel Cell Carcinoma. Clin. Chem. 2019, 65, 462–472. [Google Scholar] [CrossRef] [PubMed]
  40. Zhang, T.; Agarwal, A.; Almquist, R.G.; Runyambo, D.; Park, S.; Bronson, E.; Boominathan, R.; Rao, C.; Anand, M.; Oyekunle, T.; et al. Expression of Immune Checkpoints on Circulating Tumor Cells in Men with Metastatic Prostate Cancer. Biomark. Res. 2021, 9, 14. [Google Scholar] [CrossRef]
  41. Thompson, J.C.; Fan, R.; Black, T.; Yu, G.H.; Savitch, S.L.; Chien, A.; Yee, S.S.; Sen, M.; Hwang, W.T.; Katz, S.I.; et al. Measurement and Immunophenotyping of Pleural Fluid EpCAM-Positive Cells and Clusters for the Management of Non-Small Cell Lung Cancer Patients. Lung Cancer 2019, 127, 25–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Rebuzzi, S.E.; Rescigno, P.; Catalano, F.; Mollica, V.; Vogl, U.M.; Marandino, L.; Massari, F.; Mestre, R.P.; Zanardi, E.; Signori, A.; et al. Immune Checkpoint Inhibitors in Advanced Prostate Cancer: Current Data and Future Perspectives. Cancers 2022, 14, 1245. [Google Scholar] [CrossRef]
  43. Punekar, S.R.; Shum, E.; Grello, C.M.; Lau, S.C.; Velcheti, V. Immunotherapy in Non-Small Cell Lung Cancer: Past, Present, and Future Directions. Front. Oncol. 2022, 12, 877594. [Google Scholar] [CrossRef] [PubMed]
  44. Paz-Ares, L.G.; Ramalingam, S.S.; Ciuleanu, T.E.; Lee, J.S.; Urban, L.; Caro, R.B.; Park, K.; Sakai, H.; Ohe, Y.; Nishio, M.; et al. First-Line Nivolumab Plus Ipilimumab in Advanced NSCLC: 4-Year Outcomes From the Randomized, Open-Label, Phase 3 CheckMate 227 Part 1 Trial. J. Thorac. Oncol. 2022, 17, 289–308. [Google Scholar] [CrossRef]
  45. Acheampong, E.; Spencer, I.; Lin, W.; Ziman, M.; Millward, M.; Gray, E. Is the Blood an Alternative for Programmed Cell Death Ligand 1 Assessment in Non-Small Cell Lung Cancer? Cancers 2019, 11, 920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Ilié, M.; Szafer-Glusman, E.; Hofman, V.; Chamorey, E.; Lalvée, S.; Selva, E.; Leroy, S.; Marquette, C.H.; Kowanetz, M.; Hedge, P.; et al. Detection of PD-L1 in Circulating Tumor Cells and White Blood Cells from Patients with Advanced Non-Small-Cell Lung Cancer. Ann. Oncol. 2018, 29, 193–199. [Google Scholar] [CrossRef] [PubMed]
  47. Abdo, M.; Belloum, Y.; Heigener, D.; Welker, L.; von Weihe, S.; Schmidt, M.; Heuer-Olewinski, N.; Watermann, I.; Szewczyk, M.; Kropidlowski, J.; et al. Comparative Evaluation of PD-L1 Expression in Cytology Imprints, Circulating Tumour Cells and Tumour Tissue in Non-Small Cell Lung Cancer Patients. Mol. Oncol. 2023, 17, 13415. [Google Scholar] [CrossRef]
  48. Moran, J.A.; Adams, D.L.; Edelman, M.J.; Lopez, P.; He, J.; Qiao, Y.; Xu, T.; Liao, Z.; Gardner, K.P.; Tang, C.-M.; et al. Monitoring PD-L1 Expression on Circulating Tumor-Associated Cells in Recurrent Metastatic Non-Small-Cell Lung Carcinoma Predicts Response to Immunotherapy With Radiation Therapy. JCO Precis. Oncol. 2022, 6, e2200457. [Google Scholar] [CrossRef]
  49. Tan, Z.; Yue, C.; Ji, S.; Zhao, C.; Jia, R.; Zhang, Y.; Liu, R.; Li, D.; Yu, Q.; Li, P.; et al. Assessment of PD-L1 Expression on Circulating Tumor Cells for Predicting Clinical Outcomes in Patients with Cancer Receiving PD-1/PD-L1 Blockade Therapies. Oncologist 2021, 26, e2227–e2238. [Google Scholar] [CrossRef]
  50. Wang, Y.; Kim, T.H.; Fouladdel, S.; Zhang, Z.; Soni, P.; Qin, A.; Zhao, L.; Azizi, E.; Lawrence, T.S.; Ramnath, N.; et al. PD-L1 Expression in Circulating Tumor Cells Increases during Radio(Chemo)Therapy and Indicates Poor Prognosis in Non-Small Cell Lung Cancer. Sci. Rep. 2019, 9, 566. [Google Scholar] [CrossRef] [Green Version]
  51. Kulasinghe, A.; Kapeleris, J.; Kimberley, R.; Mattarollo, S.R.; Thompson, E.W.; Thiery, J.P.; Kenny, L.; O’Byrne, K.; Punyadeera, C. The Prognostic Significance of Circulating Tumor Cells in Head and Neck and Non-Small-Cell Lung Cancer. Cancer Med. 2018, 7, 5910–5919. [Google Scholar] [CrossRef] [Green Version]
  52. Khattak, M.A.; Reid, A.; Freeman, J.; Pereira, M.; McEvoy, A.; Lo, J.; Frank, M.H.; Meniawy, T.; Didan, A.; Spencer, I.; et al. PD-L1 Expression on Circulating Tumor Cells May Be Predictive of Response to Pembrolizumab in Advanced Melanoma: Results from a Pilot Study. Oncologist 2020, 25, e520–e527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Su, K.; Guo, L.; He, K.; Rao, M.; Zhang, J.; Yang, X.; Huang, W.; Gu, T.; Xu, K.; Liu, Y.; et al. PD-L1 Expression on Circulating Tumor Cells Can Be a Predictive Biomarker to PD-1 Inhibitors Combined with Radiotherapy and Antiangiogenic Therapy in Advanced Hepatocellular Carcinoma. Front. Oncol. 2022, 12, 873830. [Google Scholar] [CrossRef] [PubMed]
  54. Vardas, V.; Tolios, A.; Christopoulou, A.; Georgoulias, V.; Xagara, A.; Koinis, F.; Kotsakis, A.; Kallergi, G. Immune Checkpoint and EMT-Related Molecules in Circulating Tumor Cells (CTCs) from Triple Negative Breast Cancer Patients and Their Clinical Impact. Cancers 2023, 15, 1974. [Google Scholar] [CrossRef]
  55. Ikeda, M.; Koh, Y.; Teraoka, S.; Sato, K.; Oyanagi, J.; Hayata, A.; Tokudome, N.; Akamatsu, H.; Ozawa, Y.; Endo, K.; et al. Longitudinal Evaluation of PD-L1 Expression on Circulating Tumor Cells in Non-Small Cell Lung Cancer Patients Treated with Nivolumab. Cancers 2021, 13, 2290. [Google Scholar] [CrossRef] [PubMed]
  56. Zhou, Q.; Liu, X.; Li, J.; Tong, B.; Xu, Y.; Chen, M.; Liu, X.; Gao, X.; Shi, Y.; Zhao, J.; et al. Circulating Tumor Cells PD-L1 Expression Detection and Correlation of Therapeutic Efficacy of Immune Checkpoint Inhibition in Advanced Non-Small-Cell Lung Cancer. Thorac. Cancer 2023, 14, 470–478. [Google Scholar] [CrossRef] [PubMed]
  57. Janning, M.; Kobus, F.; Babayan, A.; Wikman, H.; Velthaus, J.L.; Bergmann, S.; Schatz, S.; Falk, M.; Berger, L.A.; Böttcher, L.M.; et al. Determination of PD-L1 Expression in Circulating Tumor Cells of NSCLC Patients and Correlation with Response to PD-1/PD-L1 Inhibitors. Cancers 2019, 11, 835. [Google Scholar] [CrossRef] [Green Version]
  58. Spiliotaki, M.; Neophytou, C.M.; Vogazianos, P.; Stylianou, I.; Gregoriou, G.; Constantinou, A.I.; Deltas, C.; Charalambous, H. Dynamic Monitoring of PD-L1 and Ki67 in Circulating Tumor Cells of Metastatic Non-Small Cell Lung Cancer Patients Treated with Pembrolizumab. Mol. Oncol. 2023, 17, 792–809. [Google Scholar] [CrossRef]
  59. Guibert, N.; Delaunay, M.; Lusque, A.; Boubekeur, N.; Rouquette, I.; Clermont, E.; Mourlanette, J.; Gouin, S.; Dormoy, I.; Favre, G.; et al. PD-L1 Expression in Circulating Tumor Cells of Advanced Non-Small Cell Lung Cancer Patients Treated with Nivolumab. Lung Cancer 2018, 120, 108–112. [Google Scholar] [CrossRef]
  60. Ouyang, Y.; Liu, W.; Zhang, N.; Yang, X.; Li, J.; Long, S. Prognostic Significance of Programmed Cell Death-Ligand 1 Expression on Circulating Tumor Cells in Various Cancers: A Systematic Review and Meta-Analysis. Cancer Med. 2021, 10, 7021–7039. [Google Scholar] [CrossRef]
  61. Shibata, H.; Saito, S.; Uppaluri, R. Immunotherapy for Head and Neck Cancer: A Paradigm Shift From Induction Chemotherapy to Neoadjuvant Immunotherapy. Front. Oncol. 2021, 11, 727433. [Google Scholar] [CrossRef]
  62. Cohen, E.E.W.; Soulières, D.; Le Tourneau, C.; Dinis, J.; Licitra, L.; Ahn, M.J.; Soria, A.; Machiels, J.P.; Mach, N.; Mehra, R.; et al. Pembrolizumab versus Methotrexate, Docetaxel, or Cetuximab for Recurrent or Metastatic Head-and-Neck Squamous Cell Carcinoma (KEYNOTE-040): A Randomised, Open-Label, Phase 3 Study. Lancet 2019, 393, 156–167. [Google Scholar] [CrossRef]
  63. Economopoulou, P.; Koutsodontis, G.; Avgeris, M.; Strati, A.; Kroupis, C.; Pateras, I.; Kirodimos, E.; Giotakis, E.; Kotsantis, I.; Maragoudakis, P.; et al. HPV16 E6/E7 Expression in Circulating Tumor Cells in Oropharyngeal Squamous Cell Cancers: A Pilot Study. PLoS ONE 2019, 14, e0215984. [Google Scholar] [CrossRef]
  64. Economopoulou, P.; Koutsodontis, G.; Strati, A.; Kirodimos, E.; Giotakis, E.; Maragoudakis, P.; Prikas, C.; Papadimitriou, N.; Perisanidis, C.; Gagari, E.; et al. Surrogates of Immunologic Cell Death (ICD) and Chemoradiotherapy Outcomes in Head and Neck Squamous Cell Carcinoma (HNSCC). Oral Oncol. 2019, 94, 93–100. [Google Scholar] [CrossRef]
  65. Economopoulou, P.; Kladi-Skandali, A.; Strati, A.; Koytsodontis, G.; Kirodimos, E.; Giotakis, E.; Maragoudakis, P.; Gagari, E.; Maratou, E.; Dimitriadis, G.; et al. Prognostic Impact of Indoleamine 2,3-Dioxygenase 1 (IDO1) MRNA Expression on Circulating Tumour Cells of Patients with Head and Neck Squamous Cell Carcinoma. ESMO Open. 2020, 5, e000646. [Google Scholar] [CrossRef] [PubMed]
  66. Rehman, L.U.; Nisar, M.H.; Fatima, W.; Sarfraz, A.; Azeem, N.; Sarfraz, Z.; Robles-Velasco, K.; Cherrez-Ojeda, I. Immunotherapy for Prostate Cancer: A Current Systematic Review and Patient Centric Perspectives. J. Clin. Med. 2023, 12, 1446. [Google Scholar] [CrossRef]
  67. Boudadi, K.; Suzman, D.L.; Anagnostou, V.; Fu, W.; Luber, B.; Wang, H.; Niknafs, N.; White, J.R.; Silberstein, J.L.; Sullivan, R.; et al. Ipilimumab plus Nivolumab and DNA-Repair Defects in AR-V7-Expressing Metastatic Prostate Cancer. Oncotarget 2018, 9, 28561–28571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Schott, D.S.; Pizon, M.; Pachmann, U.; Pachmann, K. Sensitive Detection of PD-L1 Expression on Circulating Epithelial Tumor Cells (CETCs) Could Be a Potential Biomarker to Select Patients for Treatment with PD-1/PD-L1 Inhibitors in Early and Metastatic Solid Tumors. Oncotarget 2017, 8, 72755–72772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Tarantino, P.; Corti, C.; Schmid, P.; Cortes, J.; Mittendorf, E.A.; Rugo, H.; Tolaney, S.M.; Bianchini, G.; Andrè, F.; Curigliano, G. Immunotherapy for Early Triple Negative Breast Cancer: Research Agenda for the next Decade. NPJ Breast Cancer 2022, 8, 23. [Google Scholar] [CrossRef]
  70. Ralli, M.; Botticelli, A.; Visconti, I.C.; Angeletti, D.; Fiore, M.; Marchetti, P.; Lambiase, A.; De Vincentiis, M.; Greco, A. Immunotherapy in the Treatment of Metastatic Melanoma: Current Knowledge and Future Directions. J. Immunol. Res. 2020, 2020, 9235638. [Google Scholar] [CrossRef] [PubMed]
  71. Hong, X.; Sullivan, R.J.; Kalinich, M.; Kwan, T.T.; Giobbie-Hurder, A.; Pan, S.; LiCausi, J.A.; Milner, J.D.; Nieman, L.T.; Wittner, B.S.; et al. Molecular Signatures of Circulating Melanoma Cells for Monitoring Early Response to Immune Checkpoint Therapy. Proc. Natl. Acad. Sci. USA 2018, 115, 2467–2472. [Google Scholar] [CrossRef] [Green Version]
  72. Chalfin, H.J.; Pramparo, T.; Mortazavi, A.; Niglio, S.A.; Schonhoft, J.D.; Jendrisak, A.; Chu, Y.L.; Richardson, R.; Krupa, R.; Anderson, A.K.L.; et al. Circulating Tumor Cell Subtypes and T-Cell Populations as Prognostic Biomarkers to Combination Immunotherapy in Patients with Metastatic Genitourinary Cancer. Clin. Cancer Res. 2021, 27, 1391–1398. [Google Scholar] [CrossRef]
  73. Kawahara, T.; Ishiguro, Y.; Ohtake, S.; Kato, I.; Ito, Y.; Ito, H.; Makiyama, K.; Kondo, K.; Miyoshi, Y.; Yumura, Y.; et al. PD-1 and PD-L1 Are More Highly Expressed in High-Grade Bladder Cancer than in Low-Grade Cases: PD-L1 Might Function as a Mediator of Stage Progression in Bladder Cancer. BMC Urol. 2018, 18, 97. [Google Scholar] [CrossRef] [Green Version]
  74. Morelli, M.B.; Amantini, C.; de Vermandois, J.A.R.; Gubbiotti, M.; Giannantoni, A.; Mearini, E.; Maggi, F.; Nabissi, M.; Marinelli, O.; Santoni, M.; et al. Correlation between High PD-L1 and EMT/Invasive Genes Expression and Reduced Recurrence-Free Survival in Blood-Circulating Tumor Cells from Patients with Non-Muscle-Invasive Bladder Cancer. Cancers 2021, 13, 5989. [Google Scholar] [CrossRef] [PubMed]
  75. Micalizzi, D.S.; Maheswaran, S.; Haber, D.A. A Conduit to Metastasis: Circulating Tumor Cell Biology. Genes. Dev. 2017, 31, 1827. [Google Scholar] [CrossRef] [PubMed]
  76. Aceto, N.; Bardia, A.; Miyamoto, D.T.; Donaldson, M.C.; Wittner, B.S.; Spencer, J.A.; Yu, M.; Pely, A.; Engstrom, A.; Zhu, H.; et al. Circulating Tumor Cell Clusters Are Oligoclonal Precursors of Breast Cancer Metastasis. Cell 2014, 158, 1110–1122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Lin, D.; Shen, L.; Luo, M.; Zhang, K.; Li, J.; Yang, Q.; Zhu, F.; Zhou, D.; Zheng, S.; Chen, Y.; et al. Circulating Tumor Cells: Biology and Clinical Significance. Signal Transduct. Target. Ther. 2021, 6, 404. [Google Scholar] [CrossRef]
  78. Steinert, G.; Schölch, S.; Niemietz, T.; Iwata, N.; García, S.A.; Behrens, B.; Voigt, A.; Kloor, M.; Benner, A.; Bork, U.; et al. Immune Escape and Survival Mechanisms in Circulating Tumor Cells of Colorectal Cancer. Cancer Res. 2014, 74, 1694–1704. [Google Scholar] [CrossRef] [Green Version]
  79. Lian, S.; Xie, X.; Lu, Y.; Jia, L. Checkpoint CD47 Function On Tumor Metastasis And Immune Therapy. Onco Targets Ther. 2019, 12, 9105–9114. [Google Scholar] [CrossRef] [Green Version]
  80. Lian, S.; Xie, R.; Ye, Y.; Lu, Y.; Cheng, Y.; Xie, X.; Li, S.; Jia, L. Dual Blockage of Both PD-L1 and CD47 Enhances Immunotherapy against Circulating Tumor Cells. Sci. Rep. 2019, 9, 4532. [Google Scholar] [CrossRef] [Green Version]
  81. Cheng, M.; Chen, Y.; Xiao, W.; Sun, R.; Tian, Z. NK Cell-Based Immunotherapy for Malignant Diseases. Cell. Mol. Immunol. 2013, 10, 230–252. [Google Scholar] [CrossRef] [Green Version]
  82. Liang, S.; Xu, K.; Niu, L.; Wang, X.; Liang, Y.; Zhang, M.; Chen, J.; Lin, M. Comparison of Autogeneic and Allogeneic Natural Killer Cells Immunotherapy on the Clinical Outcome of Recurrent Breast Cancer. Onco Targets Ther. 2017, 10, 4273–4281. [Google Scholar] [CrossRef] [Green Version]
  83. Lin, M.; Liang, S.Z.; Shi, J.; Niu, L.Z.; Chen, J.B.; Zhang, M.J.; Xu, K.C. Circulating Tumor Cell as a Biomarker for Evaluating Allogenic NK Cell Immunotherapy on Stage IV Non-Small Cell Lung Cancer. Immunol. Lett. 2017, 191, 10–15. [Google Scholar] [CrossRef]
  84. Manyam, M.; Stephens, A.J.; Kennard, J.A.; LeBlanc, J.; Ahmad, S.; Kendrick, J.E.; Holloway, R.W. A Phase 1b Study of Intraperitoneal Oncolytic Viral Immunotherapy in Platinum-Resistant or Refractory Ovarian Cancer. Gynecol. Oncol. 2021, 163, 481–489. [Google Scholar] [CrossRef] [PubMed]
  85. Yang, Y.; Qin, Z.; Du, D.; Wu, Y.; Qiu, S.; Mu, F.; Xu, K.; Chen, J. Safety and Short-Term Efficacy of Irreversible Electroporation and Allogenic Natural Killer Cell Immunotherapy Combination in the Treatment of Patients with Unresectable Primary Liver Cancer. Cardiovasc. Intervent Radiol. 2019, 42, 48–59. [Google Scholar] [CrossRef] [PubMed]
  86. Kang, Y.T.; Niu, Z.; Hadlock, T.; Purcell, E.; Lo, T.W.; Zeinali, M.; Owen, S.; Keshamouni, V.G.; Reddy, R.; Ramnath, N.; et al. On-Chip Biogenesis of Circulating NK Cell-Derived Exosomes in Non-Small Cell Lung Cancer Exhibits Antitumoral Activity. Adv. Sci. 2021, 8, 2003747. [Google Scholar] [CrossRef] [PubMed]
  87. Fu, L.Q.; Du, W.L.; Cai, M.H.; Yao, J.Y.; Zhao, Y.Y.; Mou, X.Z. The Roles of Tumor-Associated Macrophages in Tumor Angiogenesis and Metastasis. Cell. Immunol. 2020, 353, 104119. [Google Scholar] [CrossRef] [PubMed]
  88. Meng, X.; Wang, M.; Zhang, K.; Sui, D.; Chen, M.; Xu, Z.; Guo, T.; Liu, X.; Deng, Y.; Song, Y. An Application of Tumor-Associated Macrophages as Immunotherapy Targets: Sialic Acid-Modified EPI-Loaded Liposomes Inhibit Breast Cancer Metastasis. AAPS PharmSciTech 2022, 23, 285. [Google Scholar] [CrossRef] [PubMed]
  89. Stojadinovic, A.; Mittendorf, E.A.; Holmes, J.P.; Amin, A.; Hueman, M.T.; Ponniah, S.; Peoples, G.E. Quantification and Phenotypic Characterization of Circulating Tumor Cells for Monitoring Response to a Preventive HER2/Neu Vaccine-Based Immunotherapy for Breast Cancer: A Pilot Study. Ann. Surg. Oncol. 2007, 14, 3359–3368. [Google Scholar] [CrossRef] [PubMed]
  90. Kolostova, K.; Pospisilova, E.; Matkowski, R.; Szelachowska, J.; Bobek, V. Immune Activation of the Monocyte-Derived Dendritic Cells Using Patients Own Circulating Tumor Cells. Cancer Immunol. Immunother. 2022, 71, 2901–2911. [Google Scholar] [CrossRef]
Table 2. Prognostic value of PD-L1+CTCs in various types of cancers.
Table 2. Prognostic value of PD-L1+CTCs in various types of cancers.
Type of CancerCTC Isolation TechniqueCTC Detection MethodNumber of Samples (Positivity)TherapyClinical OutcomeRef.
NSCLCCellSieve Microfiltration AssayLifeTracDx PD-L1 test30 (87%); low PD-L1
30 (13%); high PD-L1
ICIsYes;
PFS-18 months
(p = 0.0112)
PFS-24 months
(p = 0.0112)
[48]
Different advanced cancersPep@MNPsIF155 (81.9%)ICIsYes;
PFS (p < 0.0001)
OS (p = 0.0235)
[49]
NSCLCGraphene oxide (GO) ChipIF and qPCR38 (69.4%)Radiation or chemoradiationYes;
5% cutoff (p = 0.017)
[50]
NSCLCCellSearchCellSearch53 (9.4%)ICIsYes;
CTC count
PFS (p = 0.006)
OS (p < 0.001)
PD-L1+CTCs
OS (p = 0.002)
[33]
NSCLCCellSearchCellSearch39 (33.3%)ICIsYes;
PFS (p = 0.040)
OS (p < 0.001)
[35]
HNSCCClearCell FX systemIF11 (54.4%)Treatment
naïve
Yes;
PFS (p = 0.0485)
[51]
HNSCCFicoll–Hypaque density gradientRT-qPCR94 (25.5%); baseline
34 (23.5%);
after IC
54 (22.2%);
at the end of treatment
ChemotherapyYes;
PFS (p = 0.001)
OS (p < 0.001)
[10]
Various types of cancerPep@MNPsIF35 (74%)PD-1 inhibitor IBI308Yes;
PFS (p = 0.002)
[49]
AMFicoll–Hypaque density gradientFlow cytometric staining25 (64%)PembrolizumabYes;
PFS (p = 0.018)
12-month PFS
(p = 0.012)
[52]
HCCCytoSorter™ BioScanner systemCytoSorter™ CTC PD-L1 Kit47 (48.9%);
<2 PD-L1+CTC
47 (51.1%);
≥2 PD-L1+CTC
PD-1 inhibitor, IMRT, antiangiogenic therapyYes;
OS (p = 0.001)
[53]
TNBCFicoll–Hypaque density gradientIF64 (41%)ChemotherapyYes;
OS (p < 0.001)
[54]
aUCCellSearchCellSearch16 (62.5%)Palliative systemic treatmentYes;
≥5 CTC
OS (p = 0.007)
[38]
Table 3. Predictive value of PD-L1+CTCs in various types of cancers.
Table 3. Predictive value of PD-L1+CTCs in various types of cancers.
Type of CancerCTC Isolation TechniqueCTC Detection MethodNumber of Samples (Positivity)TherapyResponse to TherapyRef.
NSCLCMCA systemMCA system44 (82%); baseline
31 (58%); week 4
16 (56%); week 8
13 (62%); week 12
11 (55%); week 24
ICIsYes; p < 0.05[55]
NSCLCCellSieve Microfiltration AssayLifeTracDx PD-L1 test30 (87%);
low PD-L1
30 (13%);
high PD-L1
ICIsYes;
PFS-24 months
(p = 0.0091)
OS-18 months
(p = 0.0410)
[48]
NSCLCCyttel methodIF117 (53.0%)ICIsNo;
prolonged mPFS-5.6 months
(p = 0.519)
[56]
NSCLCParsortix systemIF89 (56%);
≥1 PD-L1+CTC
89 (26%);
≥3 PD-L1+CTC
ICIsYes;
Response
(decrease or stable PD-L1+CTC)
Disease progression (increase PD-L1+CTC)
(p = 0.001)
[57]
Different advanced cancersPep@MNPsIF155 (81.9%)ICIsYes; ORR (p = 0.018) DCR (p < 0.0001)[49]
NSCLCFicoll–Hypaque density gradientIF47 (86%); baseline
43 (89%); after first cycle
23 (76%); after third cycle
19 (82%); PMR
PembrolizumabYes;
a decrease of PD-L1low CTC, partial response after the first cycle
[58]
AMFicoll–Hypaque density gradientFlow cytometric staining25 (64%)PembrolizumabYes;
PD-L1+CTCs higher in responders
(p = 0.005)
[52]
HCCCytoSorter™ BioScanner systemCytoSorter™ CTC PD-L1 Kit47 (48.9%);
<2 PD-L1+CTC
47 (51.1%);
≥2 PD-L1+CTC
PD-1 inhibitor, IMRT, antiangiogenic therapyYes;
<2 PD-L1+CTCs higher ORR
(p = 0.007)
[53]
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

Strati, A.; Economopoulou, P.; Lianidou, E.; Psyrri, A. Clinical Significance of PD-L1 Status in Circulating Tumor Cells for Cancer Management during Immunotherapy. Biomedicines 2023, 11, 1768. https://doi.org/10.3390/biomedicines11061768

AMA Style

Strati A, Economopoulou P, Lianidou E, Psyrri A. Clinical Significance of PD-L1 Status in Circulating Tumor Cells for Cancer Management during Immunotherapy. Biomedicines. 2023; 11(6):1768. https://doi.org/10.3390/biomedicines11061768

Chicago/Turabian Style

Strati, Areti, Panagiota Economopoulou, Evi Lianidou, and Amanda Psyrri. 2023. "Clinical Significance of PD-L1 Status in Circulating Tumor Cells for Cancer Management during Immunotherapy" Biomedicines 11, no. 6: 1768. https://doi.org/10.3390/biomedicines11061768

APA Style

Strati, A., Economopoulou, P., Lianidou, E., & Psyrri, A. (2023). Clinical Significance of PD-L1 Status in Circulating Tumor Cells for Cancer Management during Immunotherapy. Biomedicines, 11(6), 1768. https://doi.org/10.3390/biomedicines11061768

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